You are on page 1of 885

Hematology/Hemostasis Reference Ranges

Unless otherwise noted, data for reference range tables were compiled from multiple sources and may vary slightly from ranges listed within chapters.
Each laboratory must establish its particular ranges based on its instrumentation, methodology, and demographics of the population it serves.

Hematology Reference Ranges (Adult)


Assay Adult Male Adult Female Common Units SI Units
RBC 4.60-6.00 4.00-5.40 106/L 1012/L
HGB (Hb) 14.0-18.0 12.0-15.0 g/dL (g/L)
(140-180) (120-150)
HCT 40-54 35-49 % (L/L)
(0.40-0.54) (0.35-0.49)
MCV 80-100 80-100 fL fL
MCH 26-32 26-32 pg pg
MCHC 32-36 32-36 % g/dL
RDW 11.5-14.5 11.5-14.5 % %
Retics (automated) 25-75 25-75 103/L 109/L
Visual retics* 0.5-1.5 0.5-1.5 %
NRBCs 0 0 /100WBC /100WBC
WBCs 4.5-11.5 4.5-11.5 103/L 109/L
% ABS % ABS 103/L 109/L
SEGs (PMNs) 50-70 2.3-8.1 50-70 2.3-8.1 103/L 109/L
BANDs 0-5 0-0.6 0-5 0-0.6 103/L 109/L
ANC 2.3-8.1 2.3-8.1 103/L 109/L
LYMPHs 18-42 0.8-4.8 18-42 0.8-4.8 103/L 109/L
MONOs 2-11 0.45-1.3 2-11 0.45-1.3 103/L 109/L
EOs 1-3 0-0.4 1-3 0-0.4 103/L 109/L
BASOs 0-2 0-0.1 0-2 0-0.1 103/L 109/L
PLTs 150-450 150-450 103/L 109/L
MPV 6.8-10.2 6.8-10.2 fL fL
Erythrocyte mass <36mL/kg <32mL/kg
Erythrocyte sedimentation rate, 0-50 years: 0-15mm 0-50 years: 0-20mm
Westergren 1 hour >50 years: 0-20mm >50 years: 0-30mm

*Reticulocyte counts performed by manual methods.


HGB, Hemoglobin; HCT, hematocrit; MCV, mean cell volume; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration; RDW, red blood cell distribution width;
MPV, mean platelet volume; PLTs, platelets; PMNs, polymorphonuclear neutrophils; Segs, segmented neutrophils; Lymphs, lymphocytes; Monos, monocytes; Eos, eosinophils;
Basos, basophils; ANC, absolute neutrophil count, includes segs and bands; Retic, reticulocyte.

Tests Commonly Used to Assess Anemia (same for male and female, except noted)
Test Reference Interval
Serum Iron 50-160g/dL
Total iron-binding capacity 250-400g/dL
Percent transferrin saturation 20-55
Serum ferritin, male 15-400g/mL
Serum ferritin, female 10-106g/mL
Vitamin B12 200-850pg/mL
Serum folate 2.0-10.0g/mL
RBC folate >120g/mL
HGB A (electrophoresis) 95-100%
HGB A2 (electrophoresis) 0-3.5%
HGB F, Adult 0-2.0%
HGB F, Newborn 65-90%
Haptoglobin 31-209mg/dL
Free serum hemoglobin 0-10mg/dL

RBC, Red blood cell; HGB, hemoglobin.


Hematology Reference Ranges (Pediatric)
Common
Assay 0-1d 2-4d 5-7d 8-14d 15-30d 1-2mo 3-5mo 6-11mo 1-3y 4-7y 8-13y Units SI Units
RBC 4.10-6.10 4.36-5.96 4.20-5.80 4.00-5.60 3.20-5.00 3.40-5.00 3.65-5.05 3.60-5.20 3.40-5.20 4.00-5.20 4.00-5.40 106/L 1012/L
HGB (Hb) 16.5-21.5 16.4-20.8 15.2-20.4 15.0-19.6 12.2-18.0 10.6-16.4 10.4-16.0 10.4-15.6 9.6-15.6 10.2-15.2 12.0-15.0 g/dL (g/L)
(165-215) (164-208) (152-204) (150-196) (122-180) (106-164) (104-160) (104-156) (96-156) (102-152) (120-150)
HCT 48-68 48-68 50-64 46-62 38-53 32-50 35-51 35-51 38-48 34-48 35-49 % L/L
MCV 95-125 98-118 100-120 95-115 93-113 83-107 78-102 76-92 78-94 80-94 80-94 fL fL
MCH 30-42 30-42 30-42 30-42 28-40 27-37 25-35 23-31 23-31 23-31 26-32 pg pg
MCHC 30-34 30-34 30-34 30-34 30-34 31-36 32-36 32-36 32-36 32-36 32-36 % g/dL
RDW * * * * * * * 11.5-14.5 11.5-14.5 11.5-14.5 11.5-14.5 % %
RETICs 1.5-5.8 1.3-4.7 0.2-1.4 0-1.0 0.2-1.0 0.8-2.8 0.5-1.5 0.5-1.5 0.5-1.5 0.5-1.5 0.5-1.5 % %
NRBCs 2-24 5-9 0-1 0 0 0 0 0 0 0 0 /100WBC /100WBC
WBC 9.0-37.0 8.0-24.0 5.0-21.0 5.0-21.0 5.0-21.0 6.0-18.0 6.0-18.0 6.0-18.0 5.5-17.5 5.0-17.0 4.5-13.5 103/L 109/L
SEGs/PMNs 37-67 30-60 27-51 22-46 20-40 20-40 18-38 20-40 22-46 29-65 23-53 % %
BANDs 3-11 3-9 1-9 0-5 0-5 0-5 0-5 0-5 0-5 0-5 0-5 % %
LYMPHs 18-38 16-46 24-54 30-62 41-61 42-72 45-75 48-78 37-73 29-65 23-53 % %
MONOs 3-14 4-17 2-15 3-14 2-11 2-11 2-11 2-11 2-11 2-11 2-11 % %
EOs 1-4 1-5 2-6 1-5 1-5 1-4 1-4 1-4 1-4 1-4 1-4 % %
BASOs 0-2 0-2 0-2 0-2 0-2 0-2 0-2 0-2 0-2 0-2 0-2 % %
ANC 3.7-30 2.6-17.0 1.5-12.6 1.2-11.6 1.0-9.5 1.2-8.1 1.1-7.7 1.2-8.1 1.2-8.9 1.5-11.0 1.6-9.5 103/L 109/L
PLTs 150-450 103/L 109/L

*The RDW is markedly elevated in newborns, with a range of 14.2 to 19.9% in the first few days of life, gradually decreasing until it reaches adult levels by 6 months of age.
From Riley Hospital for Children, Indiana University Health, Indianapolis, IN.
HGB, Hemoglobin; PMNs, polymorphonuclear neutrophils; HCT, hematocrit; MCV, mean cell volume; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration;
RDW, red blood cell distribution width; Lymphs, lymphocytes; Monos, monocytes; Eos, eosinophils; ANC, absolute neutrophil count, includes segs and band; Segs, segmented
neutrophils; Retic, reticulocyte count; PLTs, platelets; Basos, basophils; NRBC, nucleated red blood cells.

Bone Marrow Aspirate


WBC Differential Range (%) Erythrocyte Series Range (%)
Blasts 0-3
Promyelocytes 1-5 Pronormoblasts 0-1
N. myelocytes 6-17 Basophilic NB 1-4
N. metamyelocytes 3-20 Polychromatophilic NB 10-20
N. bands 9-32 Orthochromic NB 6-10
N. segmented (polymorphonuclear) 7-30
Eosinophils 0-3 Other
Basophils 0-1 M:E ratio 1.5-3.3:1
Lymphocytes 5-18 Megakaryoctyes 2-10/lpf
Plasma cells 0-1
Monocytes 0-1
Histiocytes (macrophages) 0-1

N, Neutrophilic; NB, normoblast; M:E, myeloid:erythroid; lpf, low power field.


REGISTER TODAY!

To access your Student Resources, visit:

http://evolve.elsevier.com/Rodak/

Register today and gain access to:


Glossary
Provides definitions for essential hematology terms.
Weblinks
Links to places of interest on the web specifically for
hematology.
BE R N A D E T T E F . R O D A K , MS, MLS
Professor
Clinical Laboratory Science Program
Department of Pathology and Laboratory Medicine
Indiana University School of Medicine
Indianapolis, Indiana

GE O R G E A . F R I T S M A , MS, MLS
Manager
The Fritsma Factor, Your Interactive Hemostasis Resource
Birmingham, Alabama

EL A I N E M . K E O H A N E , PhD, MLS
Professor
Department of Clinical Laboratory Sciences
University of Medicine and Dentistry of New Jersey
School of Health Related Professions
Newark, New Jersey
3251 Riverport Lane
St. Louis, Missouri 63043

HEMATOLOGY: CLINICAL PRINCIPLES AND APPLICATIONS ISBN: 978-1-4377-0692-5


Copyright 2012, 2007, 2002, 1995 by Saunders, an imprint of Elsevier Inc.

Chapter 47: Coagulation Instrumentation by David L. McGlasson is Public Domain.

All rights reserved. No part of this publication may be reproduced or transmitted in any form or by
any means, electronic or mechanical, including photocopy, recording, or any information storage and
retrieval system, without permission in writing from the publisher.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic
or mechanical, including photocopying, recording, or any information storage and retrieval system,
without permission in writing from the publisher. Details on how to seek permission, further
information about the Publishers permissions policies and our arrangements with organizations such
as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website:
www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the
Publisher (other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical
treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in
evaluating and using any information, methods, compounds, or experiments described herein.
In using such information or methods they should be mindful of their own safety and the safety
of others, including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check
the most current information provided (i) on procedures featured or (ii) by the manufacturer of
each product to be administered, to verify the recommended dose or formula, the method and
duration of administration, and contraindications. It is the responsibility of practitioners, relying
on their own experience and knowledge of their patients, to make diagnoses, to determine dosages
and the best treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products,
instructions, or ideas contained in the material herein.

ISBN 978-1-4377-0692-5

Publishing Director: Andrew Allen


Managing Editor: Ellen Wurm-Cutter
Publishing Services Manager: Catherine Jackson
Senior Project Manager: David Stein
Senior Designer: Amy Buxton

Working together to grow


libraries in developing countries
Printed in China www.elsevier.com | www.bookaid.org | www.sabre.org

Last digit is the print number: 9 8 7 6 5 4 3 2


To my husband, Bob, for his continued patience,
understanding, encouragement, and love.
And to Annie, who missed her walk times
and is glad to have them back.
BFR

To my wife, Margaret, and children, Greg and Teri,


for their love, support, and loyalty.
GAF

To my students for being great teachers,


and to Camryn, Riley, Harper, Jackie, Alana, and Kenny
for reminding me about the important things in life.
EMK
This page intentionally left blank
Reviewers
Shamina Davis, MS, MT(ASCP) Amy Kapanka, MS, MT(ASCP)SC
Faculty, Bachelors of Applied Technology MLT Program Director and Professor
College of Applied Technologies and General Studies Hawkeye Community College
Office of Applied Technology Waterloo, Iowa
University of Texas at Brownsville
Brownsville, Texas E. Anne Stiene-Martin, PhD, MT(ASCP)
Professor Emeritus
David Hoblit, MD Department of Clinical and Reproductive Sciences
Faculty, Department of Pathology University of Kentucky
University of North Texas Health Science Center Lexington, Kentucky
Instructor, Department of Clinical Laboratory Science
Tarleton State University
Fort Worth, Texas

vii
Contributors

Larry D. Brace, PhD, MT(ASCP)SH Magdalena Czader, MD, PhD


Emeritus Professor of Pathology Associate Professor and Director
University of Illinois at Chicago Division of Hematopathology
Chicago, Illinois Department of Pathology and Laboratory Medicine
Scientific Director of Laboratories Director, Clinical Flow Cytometry Laboratory
Edward Hospital Indiana University School of Medicine
Naperville, Illinois Indianapolis, Indiana
Thrombocytopenia and Thrombocytosis Flow Cytometric Analysis in Hematologic Disorders
Qualitative Disorders of Platelets and Vasculature Mature Lymphoid Neoplasms

Deanne H. Chapman, BS, MT(ASCP)SH Kathryn Doig, PhD, MT(ASCP)


Technical Sales Manager Professor and Associate Dean
Bayer HealthCare LLC, Diagnostics Division Biomedical Laboratory Diagnostics
Tarrytown, New York College of Natural Science
Automated Cell-Counting Instrumentation Michigan State University
East Lansing, Michigan
Karen S. Clark, BS, MT(ASCP)SH Erythrocyte Production and Destruction
Point of Care Manager Energy Metabolism and Membrane Physiology of the Erythrocyte
Baptist Memorial Hospital Examination of the Peripheral Blood Film and Correlation with the Complete
Memphis, Tennessee Blood Count
Routine and Point-of-Care Testing in Hematology: Manual and Disorders of Iron and Heme Metabolism
Semiautomated Methods Introduction to Increased Destruction of Erythrocytes

Mary Coleman, MS, MLS(ASCP)CM, SH(ASCP)CM, Peter D. Emanuel, MD


CG(ASCP)CM Director, Winthrop P. Rockefeller Cancer Institute
Assistant Professor Professor of Medicine
Department of Pathology Kent Westbrook, MD Endowed Chair
University of North Dakota Clinical Laboratory University of Arkansas for Medical Sciences
Science Program Little Rock, Arkansas
Grand Forks, North Dakota Introduction to Leukocyte Neoplasms
Hemoglobin Metabolism
Iron Metabolism Sheila A. Finch, CHSP, CHMM, MS, BS, MT(ASCP)
System Director, Environment of Care/ISO Administrator
Leilani Collins, MS, MT(ASCP)SH Detroit Medical Center
Associate Professor Detroit, Michigan
Medical Technology Program Safety in the Hematology Laboratory
College of Allied Health Science
University of Tennessee Health Science Center
Memphis, Tennessee
Body Fluids in the Hematology Laboratory

viii
Contributors ix

George A. Fritsma, MS, MLS Mark E. Lasbury, MS, PhD


Manager Assistant Scientist
The Fritsma Factor, Your Interactive Hemostasis Resource Department of Pathology and Laboratory Medicine
Birmingham, Alabama Indiana University School of Medicine
An Overview of Clinical Laboratory Hematology Indianapolis, Indiana
Quality Assurance in Hematology and Hemostasis Testing Molecular Diagnostics in the Clinical Laboratory
Energy Metabolism and Membrane Physiology of the Erythrocyte
Platelet Production, Structure, and Function Susan J. Leclair, PhD, MLS
Bone Marrow Examination Chancellor Professor
Normal Hemostasis and Coagulation Department of Medical Laboratory Science
Hemorrhagic Coagulation Disorders University of Massachusetts Dartmouth
Thrombosis Risk Testing Dartmouth, Massachusetts
Laboratory Evaluation of Hemostasis Acute Leukemias
Monitoring Antithrombotic Therapies
Sharral Longanbach, SH(ASCP)
Margaret G. Fritsma, MA, MT(ASCP)SBB Senior Technical Application Specialist
Associate Professor, Retired Siemens Healthcare Diagnostics
School of Health Professions Deerfield, Illinois
Division of Laboratory Medicine, Department of Pathology Automated Cell-Counting Instrumentation
University of Alabama at Birmingham
Birmingham, Alabama Lynn B. Maedel, MS, MLS(ASCP)SH
Normal Hemostasis and Coagulation Executive Director
Glossary Colorado Association for Continuing Medical Laboratory
Education, Inc. (CACMLE)
Linda H. Goossen, PhD, MT(ASCP) Denver, Colorado
Professor Adjunct Faculty
Clinical Laboratory Science Michigan State University
College of Health Professions East Lansing, Michigan
Grand Valley State University Examination of the Peripheral Blood Film and Correlation with the Complete
Grand Rapids, Michigan Blood Count
Anemias Caused by Defects of DNA Metabolism
Pediatric and Geriatric Hematology David L. McGlasson, MS, MLS(ASCP)CM
Clinical Research Scientist
Teresa G. Hippel, BS, MT(ASCP)SH Clinical Research Division
Laboratory Manager Wilford Hall Medical Center
Baptist Memorial Hospital United States Air Force
Memphis, Tennessee Lackland Air Force Base, Texas
Routine and Point-of-Care Testing in Hematology: Manual and Coagulation Instrumentation
Semiautomated Methods
Rakesh P. Mehta, MD
Elaine M. Keohane, PhD, MLS Assistant Professor of Clinical Medicine
Professor Indiana University School of Medicine
Department of Clinical Laboratory Sciences Indianapolis, Indiana
University of Medicine and Dentistry of New Jersey Anemias: Red Blood Cell Morphology and Approach to Diagnosis
School of Health Related Professions Thalassemias
Newark, New Jersey
Bone Marrow Failure Martha K. Miers, MS, MBA
Intrinsic Defects Leading to Increased Erythrocyte Destruction Assistant Professor
Extrinsic Defects Leading to Increased Erythrocyte Destruction Division of Medical Education and Administration
Nonimmune Causes Vice Chair, Finance and Administration
Extrinsic Defects Leading to Increased Erythrocyte Destruction Department of Pathology
Immune Causes Vanderbilt University School of Medicine
Thalassemias Nashville, Tennessee
Automated Cell-Counting Instrumentation
x Contributors

Carole A. Mullins, MPA, CLDir(NCA), BS, MT(ASCP) Larry Smith, PhD, SH(ASCP)
Adjunct Faculty Assistant Attending Scientist, Director
Nursing and Human Services Coagulation and Hemostasis Laboratories
Southwestern Michigan College Department of Clinical Laboratories
Dowagiac, Michigan Memorial Sloan-Kettering Cancer Center
Specimen Collection New York, New York
Hematopoiesis
Keila B. Poulsen, BS, MLS(ASCP)CM H, SH
Adjunct Faculty Anne Stiene-Martin, PhD
Brigham Young University Professor Emeritus
Provo, Utah Clinical Laboratory Science Program
Hematology/Histology Supervisor Department of Clinical Science
Eastern Idaho Regional Medical Center College of Health Sciences
Idaho Falls, Idaho University of Kentucky
Cellular Structure and Function Lexington, Kentucky
Leukocyte Development, Kinetics, and Functions
Tim R. Randolph, PhD, MLS Nonmalignant Leukocyte Disorders
Associate Professor
Department of Clinical Laboratory Science Gail H. Vance, MD
Doisy College of Health Sciences Professor
Saint Louis University Indiana University School of Medicine
Saint Louis, Missouri Department of Medical and Molecular Genetics
Hemoglobinopathies (Structural Defects in Hemoglobin) Indiana UniversityPurdue University at Indianapolis
Myeloproliferative Neoplasms Clarian Health Hospitals
Staff Physician
Bernadette F. Rodak, MS, MLS Indianapolis, Carmel and Lafayette, Indiana
Professor Cytogenetics
Clinical Laboratory Science Program
Department of Pathology and Laboratory Medicine
Indiana University School of Medicine
Indianapolis, Indiana
Care and Use of the Microscope
Cytochemistry
Myelodysplastic Syndromes
Acute Leukemias
Preface

The science of clinical laboratory hematology provides for the reorganized for enhanced clarity. The discussion of white blood
analysis of normal and pathologic peripheral blood cells, cell disorders includes details of the updated World Health
hematopoietic (blood-producing) tissue, and the cells in Organization leukemia and lymphoma classifications that
cerebrospinal and serous fluid. Laboratory hematology also apply molecular techniques to detect BCR/ABL, JAK2, gene
analyzes the cells and plasma enzymes essential to clinical rearrangement, and newly described genetic markers. These
hemostasis. Although the hematology laboratory is closely chapters have been rewritten to reflect the WHO classification
aligned with clinical hematology, an internal medicine subspe- and to blend time-honored morphologic techniques with flow
cialty of oncology, it provides fundamental information for all cytometry and molecular analyses. The hemostasis chapters
patients requiring the services of the specialties of medicine now include references to emerging platelet analysis techno
and surgery. Hematology laboratory assay results help predict, logies and methods for monitoring the antiplatelet drugs
diagnose, establish the prognosis for, and monitor the treat- eptifibitide, abciximab, integrilin, clopidogrel, prasugrel, and
ment for a variety of medical disorders. Likewise hematology aspirin. The authors have updated anticoagulant monitoring
results are used to establish the need for surgery, monitor the descriptions to include chromogenic substrate analyses and
effects of surgery, and monitor treatments that support surgical new means for monitoring direct thrombin inhibitors and the
procedures. oral anticoagulants rivaroxaban, dabigatran, and apixiban, cur-
Clinical laboratory hematology has been enhanced by pro- rently in clinical trials. Sections on automated coagulometers
found changes, as reflected in the numerous updates in this, and point-of-care coagulation testing are rewritten for clarity
the fourth edition of Hematology: Clinical Principles and Applica- and timeliness.
tions. Automation and digital data management have revolu-
tionized the way blood specimens are transported and stored,
ORGANIZATION
the way laboratory assays are ordered, the way laboratory assay
results are validated and reported, and the clinical science of Hematology: Clinical Principles and Applications is organized into
laboratory result interpretation. 8 parts, 47 chapters, a detailed appendix, and a fully rewritten
Molecular diagnosis has augmented and in many instances glossary.
replaced long-indispensable laboratory assays. Hematological
disorders have been reclassified on the basis of phenotypic, Part I: Introduction to Hematology
cytogenetic, and point mutation analyses. Diagnoses that once Chapters 1 to 5 preview the science of clinical laboratory hema-
depended on the analysis of cell morphology and cytochemical tology and discuss current approaches to medical laboratory
stains now rely on flow cytometry, cytogenetic testing, fluo scientist and patient safety, peripheral blood specimen collec-
rescence in situ hybridization, end-point and real-time poly- tion, care and use of the light microscope, and hematology and
merase chain reaction assays, gene sequencing, and microarrays. hemostasis quality assurance. Updates to the quality assurance
Therapeutic regimen monitoring has shifted to the manage- chapter include discussions of new assay validation, accuracy
ment of biologic response modifiers in place of traditional and precision exercises, moving averages, lot-to-lot validations,
chemotherapy and minimal residual disease characterization clinical efficacy analyses, and receiver-operating characteristic
at the molecular and not the cellular level. Hemostasis has analyses.
grown to encompass thrombophilia testing, methods that reli-
ably monitor newly available antiplatelet and anticoagulant Part II: Hematopoiesis
drugs, molecular analysis, and a shift from clot-based to func- Chapters 6 to 13, richly illustrated with color-balanced photo-
tional and chromogenic assays. micrographs, describe the components of a hematologic cell
Some specific advances that appear in this fourth edition of and the biological and genetic details of erythropoiesis (red
Hematology: Clinical Principles and Applications include updated blood cell production), leukopoiesis (white blood cell produc-
stem cell differentiation models based on increased under- tion), and megakaryopoiesis (platelet production). Chapters 9
standing of cell signaling mechanisms and anemia classifica- to 11 examine mature red blood cell metabolism; iron metabo-
tions that incorporate updated cell membrane structure and lism; the production, structure, and function of hemoglobin;
receptors, cytochemical growth factors, and single nucleotide and red blood cell senescence and destruction. Chapter 12
polymorphism analyses. The chapters on hemolytic anemia are discusses the production of WBCs and their kinetic functions.

xi
xii Preface

Chapter 13 provides detail on platelet production, structure, Part VI: Hematology in Selected Populations
and function including adhesion, aggregation, and activation. Chapter 38, newly written, provides valuable laboratory infor-
mation on the hematology of the pediatric and geriatric
Part III: Routine Laboratory Evaluation of populations.
Blood Cells
Chapter 14 describes time-honored manual hematologic Part VII: Cell-Counting Automation
procedures that are employed daily such as manual cell counts, Chapter 39 reviews current automated hematology analyzers.
hemoglobin, and hematocrit determinations and compares
them to current point-of-care technology. Chapter 15 describes Part VIII: Hemostasis and Thrombosis
peripheral blood film preparation examination and peripheral Chapter 40 provides the coagulation cascade mechanism and
blood cell morphology. Chapter 16 follows up with bone relates it to current cell-based hemostasis models. Chapter 41
marrow aspirate and biopsy collection, preparation, examina- details hemorrhagic disorders, newly expanded to include
tion, and reporting. Chapter 17 describes the methods for management of the acute coagulopathy of trauma and shock.
analyzing the normal and pathological cells of cerebrospinal Chapter 42 describes laboratory tests that predict and monitor
fluid, joint fluid, transudates, and exudates, illustrated with thrombotic diseases of the arteries and veins. Chapters 43 and
many excellent photomicrographs. 44 describe quantitative and qualitative platelet disorders, and
Chapter 45 details laboratory assays of platelets and the coagu-
Part IV: Hematopathology: lation mechanism. Chapter 46 expands on Chapter 45 with
Erythrocyte Disorders details for monitoring antithrombotic drugs, including the
Chapter 18 provides an overview of anemia, which is the most latest oral anticoagulants and a variety of antiplatelet drugs,
common hematologic disorder, and integrates peripheral and Chapter 47 reviews automated coagulation analyzers and
blood and bone marrow morphology with red blood cell bench-top point of care instrumentation.
indices, reticulocyte counts, and interpretation of pathologic
red blood cell morphology. Chapters 19 to 21 describe disor-
READERS
ders of iron and DNA metabolism, and bone marrow failure.
Chapters 22 to 25, fully revised, provide the hemolytic anemias Hematology: Clinical Principles and Applications is written for
(anemias of shortened red blood cell life span) that are caused Medical Laboratory Scientists, Medical Laboratory Technicians,
by intrinsic or extrinsic defects. Chapters 26 and 27 provide and the faculty of Medical Laboratory Science and Medical
updates in pathophysiology and address the latest advance- Laboratory Technician educational programs. It is a valuable
ments in the diagnosis and treatment of hemoglobinopathies, study guide for pathology and hematology-oncology residents
such as sickle cell disease, and the thalassemias. and fellows and a valuable shelf reference for hematologists,
pathologists, and hematology and hemostasis laboratory
Part V: Leukocyte Disorders managers.
Chapter 28 addresses nonmalignant systemic disorders that
are reflected in the distribution of peripheral blood white
TEXTBOOK FEATURES
blood cells. These include bacterial and viral infections,
systemic disorders reflected in abnormal white blood cell Hematology: Clinical Principles and Applications is logically orga-
morphology, infectious mononucleosis, and the blood cell nized and reflects the practical clinical expertise of its authors.
effects of benign lymphoproliferative disorders. There are The text is enhanced by full-color, color-balanced digital
many new, color-balanced digital photomicrographs. Chapter photomicrographs, figures and line art, and detailed text boxes
29 introduces the pathologic mechanisms of white blood and tables. Figures and tables are enhanced with stand-alone
cell neoplasms (malignancy) and Chapters 30 to 33 provide captions. Pedagogy is supported by learning objectives, real-life
updated details on time-honored but still effective cyto introductory case studies with open-ended discussion ques-
chemistry; cytogenetic procedures, which now include fluores- tions, comprehensive summaries, and study questions at the
cence in situ hybridization; and molecular diagnostics, with end of each chapter. Students preparing for examinations may
emphasis on end-point and real-time polymerase chain use the learning objectives, review questions, and chapter sum-
reaction, microarrays, and gene sequencing. Chapter 33 maries to guide their test preparation. The appendix provides
describes the technology of flow cytometry and its diagnostic examples of material safety data sheets, answers to the case
applications, includes the detection of current and emerging study questions and review questions, and an extensive, fully
phenotypic markers. The chapter provides numerous exam- updated glossary.
ples of scatterplot results in normal and leukemic conditions.
Chapters 34 to 37 provide the latest worldwide classifica-
NEW TO THIS EDITION
tions and pathophysiologic models for myeloproliferative
neoplasms, myelodysplastic syndromes, acute lymphoblastic Editor Bernadette Rodak, MS, MLS Indiana University
and myeloblastic leukemias, chronic lymphocytic leukemia, School of Medicine, who has been the lead editor for the
and solid tumor lymphoid neoplasms such as lymphoma and first three editions, and George A. Fritsma, MS, MLS,
myeloma. University of Alabama at Birmingham Department of
Preface xiii

Pathology, co-editor of the third edition, are joined for the A bulleted summary at the end of each chapter that provides
fourth edition by Elaine M. Keohane, PhD, MLS, University a comprehensive review of essential material.
of Medicine and Dentistry of New Jersey Department of Review questions at the end of each chapter that are written
Clinical Laboratory Sciences. to match the chapter objectives; they are given in the format
Co-editor George A. Fritsma, proprietor of the blog The used by certification examinations.
Fritsma Factor, Your Interactive Hemostasis Reference, spon-
sored by Precision BioLogic, Dartmouth, Nova Scotia,
rewrote Chapter 5, Quality Assurance in Hematology and
EVOLVE ANCILLARIES
Hemostasis Testing; and Chapter 16, Bone Marrow Exami-
nation. Prof. Fritsma continues as editor of Part VIII: Hemo- For the Instructor
stasis and Thrombosis. Test bank: An ExamView test bank of 850 multiple-choice
Chapter 47, Coagulation Instrumentation, has been com- questions features rationales, cognitive levels, and page
pletely updated by internationally recognized hemostasis number references to the text. Available for in-class review
researcher David McGlasson, Wilford Hall USAF Hospital, or for test development.
San Antonio, Texas. Instructors manual: The instructors manual includes one
Substantial updates were made to Chapter 29, Introduction chapter for every chapter in the text and contains key terms,
to Leukocyte Neoplasms, by Peter D. Emanuel, MD, Direc- objectives, outlines, and study questions. This can be used
tor, Winthrop P. Rockefeller Cancer Institute, Professor of in preparation for classes and during lecture.
Medicine, Kent Westbrook, MD Endowed Chair, University Case studies: 54 case studies feature photomicrographs,
of Arkansas for Medical Sciences, Little Rock, Arkansas. images, and discussion questions. These can be used in class
Dr. Keohane provided substantial updates to Chapters 23 for review of material, for practical exams, or for students
to 25, on the Hemolytic Anemias. class presentations.
Substantial updates in Chapter 20, Anemias Caused by PowerPoint presentations: One PowerPoint presentation
Defects of DNA Metabolism, and Chapter 38, Pediatric and per chapter can be used as is or as a template to prepare
Geriatric Hematology, by Linda H. Goossen, PhD, MT lectures.
(ASCP), College of Health Professions, Grand Valley State Image Collection: All of the images from the book are avail-
University, Grand Rapids, Michigan. able as electronic files that can be downloaded into Power-
Point presentations. These can be used during lecture to
illustrate important concepts.
PEDAGOGY
For the Student
Each chapter contains: Glossary: The glossary is available on the Evolve site and
Learning objectives for all cognitive domains. can be used as a quick reference to look up unfamiliar terms
One or two case studies at the beginning of each chapter electronically.
that pique the readers interest and provide open-ended Weblinks: Links to places of interest on the web specifically
discussion questions. for hematology.
Acknowledgments

Editors Elaine Keohane and George Fritsma respectfully Roslyn McQueen, Rakesh Mehta, Martha Miers, Carole Mullins,
acknowledge Bernadette F. (Bunny) Rodak, who first under- Martha Payne, Keila Poulsen, Tim Randolph, Vishnu Reddy,
stood the need for an accessible clinical laboratory hematology Larry Smith, Anne Stiene-Martin, Gail Vance, and Karen
text in 1992. Through Bunnys perseverance, Hematology: Clini- Bourlier Waldron.
cal Principles and Applications has grown to a worldwide educa- We also express personal appreciation to Ellen Wurm-
tional resource and reference for pathology and hematology Cutter, Elsevier managing editor, who has responsibly and pro-
practitioners, residents, and fellows; medical laboratory scien- fessionally managed production through three editions. Ellens
tists; and medical laboratory science students. Now in this, its professional support, prompts, and nudges kept the project on
fourth edition, the Rodak hematology text continues to lead track. We are grateful to David Stein, Catherine Jackson, and
in its contributions to clinical laboratory science and labora- Janet Lincoln for their valued editorial assistance.
tory and clinical hematology. Finally, we acknowledge the contributions of newly invited
The editors acknowledge the authors who have made and fourth edition authors, fourth edition editor Elaine Keohane,
continue to make significant contributions. These include authors who contributed manuscripts to previous editions, and
Kathryn Doig, co-editor of the third edition; Ann Bell, Larry our friends and professional colleagues who have encouraged
Brace, Carol Bradford, Sarah Burns, Michelle Butina, the late this project through the years.
Deanne Chapman, Karen Clark, Mary Coleman, Leilani Collins,
Magdalena Czader, Peter Emanuel, Sheila Finch, Margaret
Fritsma, Linda Goossen, John Griep, Teresa Hippel, Cynthia Bernadette F. Rodak, MS, MLS
Johns, John Krause, Mark Lasbury, Susan Leclair, Sharral George A. Fritsma, MS, MLS
Longanbach, Lynn Maedel, Dave McGlasson, Marisa Marques, Elaine M. Keohane, PhD, MLS

xiv
Contents

PART I Introduction to Hematology PART III Routine Laboratory Evaluation of


Blood Cells
CHAPTER 1 An Overview of Clinical Laboratory Hematology, 1
George A. Fritsma CHAPTER 14 Routine and Point-of-Care Testing in Hematology:
Manual and Semiautomated Methods, 172
CHAPTER 2 Safety in the Hematology Laboratory, 7 Karen S. Clark and Teresa G. Hippel
Sheila A. Finch
CHAPTER 15 Examination of the Peripheral Blood Film and
CHAPTER 3 Specimen Collection, 18 Correlation with the Complete Blood Count, 192
Carole A. Mullins Lynn B. Maedel and Kathryn Doig

CHAPTER 4 Care and Use of the Microscope, 32 CHAPTER 16 Bone Marrow Examination, 210
Bernadette F. Rodak George A. Fritsma

CHAPTER 5 Quality Assurance in Hematology and Hemostasis CHAPTER 17 Body Fluids in the Hematology Laboratory, 226
Testing, 41 Leilani Collins
George A. Fritsma

PART IV Hematopathology:
PART II Hematopoiesis Erythrocyte Disorders

CHAPTER 6 Cellular Structure and Function, 57 CHAPTER 18 Anemias: Red Blood Cell Morphology and
Keila B. Poulsen Approach to Diagnosis, 241
Rakesh P. Mehta
CHAPTER 7 Hematopoiesis, 66
Larry Smith CHAPTER 19 Disorders of Iron and Heme Metabolism, 253
Kathryn Doig
CHAPTER 8 Erythrocyte Production and Destruction, 86
Kathryn Doig CHAPTER 20 Anemias Caused by Defects of DNA
Metabolism, 268
CHAPTER 9 Energy Metabolism and Membrane Physiology of Linda H. Goossen
the Erythrocyte, 103
Kathryn Doig and George A. Fritsma CHAPTER 21 Bone Marrow Failure, 283
Elaine M. Keohane
CHAPTER 10 Hemoglobin Metabolism, 115
Mary Coleman CHAPTER 22 Introduction to Increased Destruction of
Erythrocytes, 299
CHAPTER 11 Iron Metabolism, 126 Kathryn Doig
Mary Coleman
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte
CHAPTER 12 Leukocyte Development, Kinetics, and Destruction, 314
Functions, 134 Elaine M. Keohane
Anne Stiene-Martin
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte
CHAPTER 13 Platelet Production, Structure, and Function, 152 DestructionNonimmune Causes, 337
George A. Fritsma Elaine M. Keohane

xv
xvi Contents

CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte


DestructionImmune Causes, 353
PART VII Cell-Counting Automation
Elaine M. Keohane
CHAPTER 39 Automated Cell-Counting Instrumentation, 598
Sharral Longanbach, Deanne H. Chapman, and
CHAPTER 26 Hemoglobinopathies (Structural Defects in
Martha K. Miers
Hemoglobin), 366
Tim R. Randolph

CHAPTER 27 Thalassemias, 390


PART VIII Hemostasis and Thrombosis
Rakesh P. Mehta and Elaine M. Keohane
CHAPTER 40 Normal Hemostasis and Coagulation, 626
Margaret G. Fritsma and George A. Fritsma
PART V Leukocyte Disorders
CHAPTER 41 Hemorrhagic Coagulation Disorders, 647
George A. Fritsma
CHAPTER 28 Nonmalignant Leukocyte Disorders, 408
Anne Stiene-Martin
CHAPTER 42 Thrombosis Risk Testing, 668
George A. Fritsma
CHAPTER 29 Introduction to Leukocyte Neoplasms, 427
Peter D. Emanuel
CHAPTER 43 Thrombocytopenia and Thrombocytosis, 694
Larry D. Brace
CHAPTER 30 Cytochemistry, 436
Bernadette F. Rodak
CHAPTER 44 Qualitative Disorders of Platelets and
Vasculature, 718
CHAPTER 31 Cytogenetics, 445
Larry D. Brace
Gail H. Vance

CHAPTER 45 Laboratory Evaluation of Hemostasis, 734


CHAPTER 32 Molecular Diagnostics in the Clinical
George A. Fritsma
Laboratory, 461
Mark E. Lasbury
CHAPTER 46 Monitoring Antithrombotic Therapies, 765
George A. Fritsma
CHAPTER 33 Flow Cytometric Analysis in Hematologic
Disorders, 488
CHAPTER 47 Coagulation Instrumentation, 783
Magdalena Czader
David L. McGlasson
CHAPTER 34 Myeloproliferative Neoplasms, 508
Tim R. Randolph
APPENDIX
CHAPTER 35 Myelodysplastic Syndromes, 533
Material Safety Data Sheet, 796
Bernadette F. Rodak

Answers, 801
CHAPTER 36 Acute Leukemias, 546
Susan J. Leclair and Bernadette F. Rodak
Glossary, 816
CHAPTER 37 Mature Lymphoid Neoplasms, 558
Magdalena Czader

PART VI Hematology in Selected Populations

CHAPTER 38 Pediatric and Geriatric Hematology, 580


Linda H. Goossen
PART I Introduction to Hematology
CHAPTER 1 An Overview of Clinical Laboratory Hematology 1

An Overview of Clinical
Laboratory Hematology
1
George A. Fritsma

T
he average human possesses 5L of blood. Blood transports oxygen from lungs to tissues; clears
OUTLINE
tissues of carbon dioxide; transports glucose, proteins, and fats; and moves wastes to the liver
History and kidneys. The liquid portion is plasma, which, among many other components, provides
Red Blood Cells coagulation enzymes that protect vessels from trauma and maintain the circulation.
Hemoglobin, Hematocrit, Plasma transports and nourishes blood cells. There are three families of blood cells: red blood
and Red Blood Cell cells (RBCs), or erythrocytes; white blood cells (WBCs), or leukocytes; and platelets, or thrombocytes.1
Indices Hematology is the study of blood cells. By expertly staining, counting, analyzing, and recording the
Reticulocytes
appearance, phenotype, and genotype of all three types of cells, the medical laboratory scientist is able
White Blood Cells
to predict, detect, and diagnose blood diseases and many systemic diseases that affect blood cells.
Platelets
Complete Blood Count Physicians rely on hematology laboratory test results to select and monitor therapy for these
Blood Film Examination disorders.
Endothelial Cells
Coagulation
HISTORY
Advanced Hematology
Procedures Early scientists such as Athanasius Kircher in 1657 described worms in the blood, and Anton van
Additional Hematology Leeuwenhoek in 1674 gave an account of RBCs,2 but it was not until the late 1800s that Giulio
Procedures Bizzozero described platelets as petites plaques.3 The development of Wright stain by James Homer
Hematology Quality Wright in 1902 opened a new world of visual blood examination through the microscope. Although
Assurance and Quality
many automated instruments now differentiate and enumerate blood cells, Wrights Romanowsky-
Control
type stain (polychromatic, a mixture of acidic and basic dyes), and refinements thereof, remains the
heart of blood cell identification.4
In the present-day hematology laboratory, RBC, WBC, and platelet appearance is analyzed visually
using 500 to 1000 light microscopy examination of cells fixed to a glass microscope slide and stained
with Wright or Wright-Giemsa stain (see Chapter 15). The scientific term for cell appearance is morpho
logy, which encompasses cell color, size, shape, cytoplasmic inclusions, and nuclear condensation.

RED BLOOD CELLS


RBCs are anucleate biconcave cells filled with a reddish protein, hemoglobin (Hb, HGB), which trans-
ports oxygen and carbon dioxide (see Chapter 10). RBCs appear pink to red and measure 6 to 8m
in diameter with a zone of pallor covering one third of their center (Figure 1-1, A), reflecting their
biconcavity (see Chapters 8 and 9).
Since before 1900, physicians and medical laboratory scientists counted RBCs in measured volumes
to detect anemia or polycythemia. Anemia means loss of oxygen-carrying capacity and is often reflected
in a reduced RBC count (see Chapters 14 and 18). Polycythemia means an increased RBC count reflect-
ing increased body RBC mass, a condition that leads to hyperviscosity (see Chapter 34). To count
RBCs, laboratory scientists carefully pipetted a tiny aliquot of whole blood and mixed it with 0.85%
(normal) saline. This saline concentration matches the osmolality of normal blood; consequently, the
suspended RBCs closely retained their native morphology, neither swelling nor shrinking. A 1:200
dilution was typical for RBC counts, and a glass pipette designed to provide this dilution, the Thoma
pipette, was used routinely until the advent of automation and is still available from clinical labora-
tory supply companies.

1
2 PART I Introduction to Hematology

standard and is mathematically converted to hemoglobin con-


centration. Drabkin reagent is used in manual and most auto-
B mated applications, although some automated hematology
profiling instruments use a formulation of the ionic surfactant
C (detergent) sodium dodecyl sulfate to reduce environmental
D
cyanide.
Hematocrit is the ratio of the volume of RBCs to the volume
of whole blood and is determined by transferring blood to a
A graduated plastic tube, centrifuging, measuring the column of
F RBCs, and dividing by the total length of RBCs plus plasma.
The normal ratio approaches 50% (see inside front cover for
reference ranges). Hematocrit is also called packed cell volume
(PCV), the packed cells referring to RBCs. Often one can see a
E G light-colored layer between the RBCs and plasma. This is the
buffy coat and contains WBCs and platelets. The laboratory
H scientist may use the three numerical results, RBC count,
hemoglobin, and hematocrit, to compute the RBC indices
Figure 1-1 A, Normal erythrocyte (red blood cell, RBC). B, Normal poly-
mean cell volume (MCV), mean cell hemoglobin (MCH), and
morphonuclear neutrophil (PMN, segmented neutrophil, seg). C, Normal
mean cell hemoglobin concentration (MCHC, see Chapter 14).
band neutrophil (band). D, Normal eosinophil (EO). E, Normal basophil (baso).
F, Normal lymphocyte (lymph). G, Normal monocyte (mono). H, Normal The MCV, although a measure of volume, reflects RBC dia
platelet. meter on a Wright-stained blood film; the MCHC reflects RBC
staining intensity or degree of pallor. The MCH expresses the
mass of hemoglobin and closely reflects the MCHC. A fourth
RBC index, RBC distribution width (RDW), expresses the
degree of variation in RBC volume. Extreme RBC volume vari-
The diluted blood was transferred to a counting chamber or ability is visible on the Wright-stained blood film as variation
hemacytometer (see Figure 14-1). The medical laboratory sci- in diameter and is called anisocytosis. The RDW is based on the
entist observed and counted RBCs in selected areas of the standard deviation of RBC volume and is routinely reported by
hemacytometer, applied a mathematical formula based on automated cell counters but cannot be provided using manual
the dilution and on the area of the hemacytometer counted RBC measurements. In addition to aiding diagnosis, the RBC
(see Chapter 14), and reported the RBC count in cells per indices provide stable measurements for internal quality
microliter (mcL), milliliter (mL, also called cubic centimeter, or control (see Chapter 5).
cc), or liter (L). Laboratory scientists routinely use 1000 visual examina-
Visual RBC counting was developed before 1900 and, tion (see Chapter 4) to review RBC morphology, commenting
although never accurate, was the only way to count RBCs until consistently on RBC diameter, color or hemoglobinization,
1958, when automated particle counters became available in shape, and the presence of cytoplasmic inclusions (see Chapter
the clinical laboratory. The first electronic counter, patented in 18). All these parametersRBC count, hemoglobin, hemato-
1953 by Joseph and Wallace Coulter of Chicago, Illinois, was crit, indices, and RBC morphologyare used to detect, diag-
used so widely that today automated cell counters are often nose, assess the severity of, and monitor the treatment of
called Coulter counters, although many high-quality competi- anemia, polycythemia, and numerous systemic conditions
tors exist (see Chapter 39).5 The Coulter principle of direct that affect RBCs. Automated hematology profiling instruments
current electrical impedance is still used for RBC counting in are used in nearly all laboratories to generate these data,
many automated hematology profiling instruments. Happily, although examination of the Wright-stained blood film is still
the widespread availability of automated cell counters has essential.
replaced visual RBC counting.
Reticulocytes
Hemoglobin, Hematocrit, and Red Blood In the Wright-stained film, 1% to 2% of RBCs exceed the 6- to
Cell Indices 8-m average diameter and stain slightly blue-gray. These are
RBCs also are assayed for hemoglobin concentration and polychromatophilic erythrocytes, newly released from the RBC
hematocrit (Hct, see Chapters 10 and 14). Hemoglobin mea- production site, the bone marrow (see Chapters 8 and 16).
surement relies on a weak solution of potassium cyanide and Polychromatophilic erythrocytes are closely observed because
potassium ferricyanide, called Drabkin reagent. An aliquot of they indicate bone marrow regeneration during blood loss and
whole blood is mixed with a measured volume of Drabkin certain anemias (see Chapters 23 to 25).
reagent, hemoglobin is converted to stable cyanmethemoglobin Methylene blue dyes, called nucleic acid stains or vital stains,
(hemiglobincyanide), and the solution is placed in a photom- are used to differentiate and count these young RBCs. Vital
eter with incident light at 540 nm wavelength. The color inten- stains are dyes absorbed by live cells. Young RBCs contain
sity is compared with that of a reagent blank and a known ribonucleic acid (RNA) and are called reticulocytes when the
CHAPTER 1 An Overview of Clinical Laboratory Hematology 3

RNA is highlighted using vital stains. Reticulocyte counting was Eosinophils (EOs; see Figure 1-1, D). Eosinophils are cells
(and remains) a tedious and imprecise visual chore until the with bright orange, regular cytoplasmic granules filled
development of automated reticulocyte counting by the TOA with antihistamine. An elevated eosinophil count is called
Corporation (presently Sysmex Corporation, Kobe, Japan) in eosinophilia and signals a response to allergy or parasitic
1990. Now all fully automated profiling instruments provide infection.
an absolute reticulocyte count and a particularly sensitive measure Basophils (basos; see Figure 1-1, E). Basophils are cells with
of bone marrow function, the immature reticulocyte count or dark purple, irregular cytoplasmic granules that obscure the
immature reticulocyte fraction. To everyones regret, it is still nucleus. An elevated basophil count is basophilia. Basophilia
necessary to confirm instrument counts visually from time to is rare and often signals a hematologic disease, such as
time, which requires that medical laboratory scientists retain leukemia.
this skill. Segs, bands, eosinophils, and basophils are collectively
called granulocytes because of their prominent cytoplasmic
granules, although their functions differ. The distribution
of eosinophils and basophils in blood is so small compared
WHITE BLOOD CELLS
with that of PMNs that the terms eosinopenia and basopenia
WBCs, or leukocytes, are not really blood cells; they are a are theoretical and unused. Leukemia is uncontrolled pro-
loosely related grouping of cell families dedicated to protecting liferation of WBCs. Leukemia may be chronic, for example
their host from infection and injury (see Chapters 12 and 28). chronic myelogenous (granulocytic) leukemia, or acute, such
WBCs hitch a ride in the blood from their source, usually as acute myeloblastic leukemia. There are several forms of
bone marrow or lymphoid tissue, to their tissue destination. granulocytic leukemias (see Chapters 34 to 36), categorized
They are so named because they are nearly colorless in an by their respective genetic aberrations (see Chapters 31 and
unstained cell suspension. In chronic leukemia, an extreme 32), and laboratory scientists are responsible for their iden-
increase in the WBC count imparts a milky appearance to the tification using Wright-stained bone marrow smears, cyto-
blood (see Chapters 34 and 36). chemical stains, cytogenetics, and molecular diagnostic
WBCs may be counted visually using a microscope, hema- technology (see Chapters 16, 30, 31, and 32).
cytometer, and a Thoma pipette. The technique is the same as Lymphocytes (lymphs, see Figure 1-1, F). Lymphocytes
RBC counting, but the typical dilution is 1:20, and the diluent comprise a complex system of cells that provide for host
is composed of dilute acetic acid in normal saline. The acid immunity. Lymphocytes recognize foreign antigens and
causes RBCs to lyse or rupture; without it, RBCs would obscure mount antibody (humoral) and cell-mediated antagonistic
the WBCs, whose count ranges from 4500 to 11,500/mcL. responses. On a Wright-stained film, most lymphocytes are
Visual WBC counting has been largely replaced by automated nearly round, are slightly larger than RBCs, and have round
hematology profiling instruments, but is accurate and useful featureless nuclei and a thin rim of nongranular cytoplasm.
in situations in which no automation is available. Laboratory An increase in the lymphocyte count is called lymphocytosis
scientists who analyze body fluids such as cerebrospinal fluid and often is associated with viral infections. An abnormally
employ visual WBC counting every day. low lymphocyte count is lymphopenia or lymphocytopenia and
A decreased WBC count (fewer than 4500/mcL) is called is associated with long-term drug therapy or immunodefi-
leukopenia, and an increased WBC count (more than 11,500/ ciency. Lymphocytes are also implicated in leukemia; chronic
mcL) is called leukocytosis, but the WBC count alone has modest lymphocytic leukemia is prevalent in people older than 70
clinical value. The laboratory scientist must differentiate the years, whereas acute lymphoblastic leukemia is the most
families of WBCs transported in the blood by using a Wright- common form of childhood leukemia (see Chapters 36 to
stained blood film and light microscopy (see Chapter 15). The 38). Laboratory scientists classify lymphocytic leukemias, of
types of WBCs are as follows: which there are several, largely based on Wright-stained
Polymorphonuclear neutrophils (PMNs, or segmented neu- blood films and flow cytometry (see Chapter 33).
trophils or segs; see Figure 1-1, B). Segs are phagocytic Monocytes (monos, see Figure 1-1, G). The mono is an
cells whose sole purpose is to engulf and destroy bacteria immature macrophage passing through the blood from its
that have been earlier labeled as harmful by the immune point of origin, usually the bone marrow, to a targeted
system. An increase in segs is called neutrophilia and often tissue location. Macrophages are the most abundant cell in
signals bacterial infection. A decrease is called neutropenia the body, more abundant than RBCs or skin cells, although
and often is caused by long-term drug administration or a they are a minor component of the blood film differential
viral infection. count. They occupy every body cavity; some are motile and
Band neutrophils (bands; see Figure 1-1, C). Bands are part some immobilized. Their task is to identify and phagocytose
of the seg family; they are simply less differentiated or (engulf) foreign particles and assist the lymphocytes in
less mature than segs. A band increase also signals bacte- mounting an immune response through the assembly and
rial infection and is customarily called a left shift. The presentation of immunogenic epitopes. On a Wright-stained
cytoplasm of segmented neutrophils and bands contains film, monos have a slightly larger diameter than other
submicroscopic, pink-staining granules filled with bacteri- WBCs, gray cytoplasm, and a lobulated nucleus. An increase
cidal secretions. in the number of monocytes may signal a hematologic
4 PART I Introduction to Hematology

disease, such as leukemia, and is called monocytosis. Scien-


COMPLETE BLOOD COUNT
tists seldom document a decreased mono count, so
the theoretical term monocytopenia is seldom used. The The medical laboratory scientist may collect a blood specimen
monocyte-macrophage cell line is another source for leuke- for the complete blood count (CBC); a phlebotomist, nurse,
mias, for instance, chronic or acute monocytic leukemia physician, or patient care technician may also collect the speci-
(see Chapter 36). men (see Chapters 3, 14, and 45). In all cases, the scientist is
responsible for the integrity of the specimen and ensures that
it is free of clots, hemolysis, and inappropriate anticoagulant-
PLATELETS
to-specimen ratios known as short draws. The medical labora-
Platelets, or thrombocytes, are true blood cells that maintain tory scientist also ensures that the specimen is fresh enough for
blood vessel integrity by instigating vessel wall repairs (see accurate analysis (see Chapter 5) and then accurately registers
Chapter 13). Platelets rapidly adhere to the surfaces of damaged the specimen in the work list, a process known as specimen
blood vessels, form aggregates with neighboring platelets to accession. Accession may be automated, relying on bar code or
plug the vessels, and secrete proteins and small molecules that radio-frequency identification technology.
trigger thrombosis, or clot formation. Platelets are the cells that Although all laboratory scientists are equipped to perform
control hemostasis, a series of cellular and plasma-based mecha- visual RBC, WBC, and platelet counts using dilution pipettes,
nisms that seals wounds, repairs vessel walls, and maintains hemacytometers, and microscopes, most laboratories employ
vascular patency. Platelets are only 2 to 4m in diameter, profiling instruments to generate the CBC parameters listed in
round or oval, anucleate, and slightly granular (see Figure 1-1, Box 1-1. The more sophisticated profiling instruments provide
H). Their diminutive size makes them appear insignificant, but comments on RBC, WBC, and platelet morphology (see
they are essential to life and are extensively studied for their Chapter 39). When one of the results from the profiling instru-
complex physiology. Uncontrolled platelet and hemostatic ment is abnormal, the instrument provides an indication of
activation is responsible for deep vein thrombosis, pulmonary this, sometimes called a flag. In this case, the scientist performs
emboli, acute myocardial infarctions (heart attacks), cerebro- a reflex blood film examination (see Chapter 15).
vascular accidents (strokes), peripheral artery disease, and The blood film examination is a specialized, demanding,
repeated spontaneous abortions (miscarriage). and fundamental CBC activity. Nevertheless, if all profiling
The medical laboratory professional counts platelets using
the same technique used in counting WBCs on a hemacyto
meter, although the counting is usually confined to the center BOX 1-1 Measurements Generated by Automated
square millimeter of the hemacytometer. Owing to their minus- Hematology Profiling Instruments
cule volume, platelets are hard to distinguish visually in a
hemacytometer, and phase microscopy provides for easier iden- RBC Parameters
tification (see Chapter 4). Automated profiling instruments RBC count
have largely replaced visual platelet counting and provide Hb
greater accuracy (see Chapter 39). Hct
One advantage of automated profiling instruments is their MCV
ability to generate a mean platelet volume (MPV), which is MCH
unavailable through visual methods. The presence of predomi- MCHC
nantly larger platelets generates an elevated MPV value, which RDW
sometimes signals a regenerative bone marrow response to Retic
platelet consumption (see Chapters 13 and 43).
Elevated platelet counts, called thrombocytosis, signal inflam- WBC Parameters
mation or trauma but carry small intrinsic significance. Essen- WBC count
tial thrombocythemia is a rare malignant condition characterized Seg count: % and absolute
by extremely high platelet counts and uncontrolled platelet Band count: % and absolute
production. Essential thrombocythemia is a life-threatening Lymph count: % and absolute
hematologic disorder (see Chapter 34). Mono count: % and absolute
A low platelet count, called thrombocytopenia, is a common EO and baso counts: % and absolute
consequence of drug treatment and may be life-threatening.
Because the platelet is responsible for normal blood vessel Platelet Parameters
maintenance and repair, thrombocytopenia is usually accom- Platelet count
panied by easy bruising and uncontrolled hemorrhage (see MPV
Chapter 43). Thrombocytopenia accounts for the majority
of hemorrhage-related emergency department visits. Accurate baso, Basophil; EO, eosinophil; Hb, hemoglobin; Hct, hematocrit; Lymph, lympho-
cyte; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration;
platelet counting is essential to patient safety for diagnosis MCV, mean cell volume; Mono, monocyte; MPV, mean platelet volume; Seg,
of thrombocytopenia in many disorders or therapeutic segmented neutrophil; RBC, red blood cell; RDW, RBC distribution width; Retic,
regimens. reticulocyte; WBC, white blood cell.
CHAPTER 1 An Overview of Clinical Laboratory Hematology 5

instrument results are normal, the blood film examination is exert control over the coagulation mechanism, and a third
omitted from the CBC. Indeed, the blood film examination is system of enzymes and cofactors digests clots to restore
waived in 50% to 80% of cases, depending on the facilitys case vessel patency, a process called fibrinolysis. Bleeding and clot-
mix. Acute care facilities report more normal CBCs than spe- ting disorders are manifold and complex, and the coagulation
cialized or tertiary care facilities, in which the percentage of section of the hematology laboratory provides a series of
blood film examinations may exceed 50%. Physicians may plasma-based laboratory assays reflecting complex interactions
request a blood film examination on the basis of clinical sus- of hematologic cells with plasma proteins (see Chapters 40
picion even when the profiling instrument results are within and 45).
normal limits (WNL). The medical laboratory scientist focuses especially on blood
specimen integrity for the coagulation laboratory, because
minor blood specimen defects, including clots, hemolysis,
BLOOD FILM EXAMINATION
lipemia, plasma bilirubin, and short draws, render the speci-
To accomplish a blood film examination, the medical labora- men useless (see Chapters 3 and 45). High-volume coagulation
tory scientist prepares a wedge-prep blood film on a glass tests suited to the acute care facility include the platelet count
microscope slide, allows it to dry, and fixes and stains it using and MPV as described earlier, prothrombin time and partial
Wright or Wright-Giemsa stain (see Chapter 15). The scientist thromboplastin time (or activated partial thromboplastin time),
affixes the slide to the microscope stage and examines the RBCs thrombin time (or thrombin clotting time), fibrinogen assay,
and platelets for abnormalities of shape, diameter, color, or and D-dimer assay (see Chapter 45). The prothrombin time
inclusions using the 50 or 100 oil immersion lens to generate and partial thromboplastin time are particularly high-volume
500 or 1000 magnification (see Chapter 4). The WBC count assays. These tests assess each arm of the coagulation pathway
and platelet count are estimated for comparison with the instru- for deficiencies and are used to monitor anticoagulant therapy.
ment counts, and all abnormalities are carefully recorded. The Another 30 to 40 low-volume assays are available in special-
scientist systematically reviews, identifies, and tabulates 100 ized or tertiary care facilities. The specialized or tertiary care
(or more) WBCs to determine their percent distribution. This coagulation laboratory with its interpretive complexities is a
process is referred to as determining the WBC differential haven for advanced medical laboratory scientists with special-
(diff). This individualized activity is highly reliant on the ized knowledge and communication skills.
scientists skill, visual acuity, and integrity, and it provides
extensive diagnostic information. Medical laboratory scientists
ADVANCED HEMATOLOGY PROCEDURES
pride themselves on their technical and analytical skills in
performing the blood film examination. The results of the CBC, Besides performing the CBC, the hematology laboratory pro-
including all profiling and blood film examination parameters vides bone marrow examinations, flow cytometry, cytogenetic analy-
and interpretive comments, are provided on a single page or sis, and molecular diagnosis assays. Performing these tests may
computer screen for physician review with abnormal results require advanced preparation or particular dedication by
highlighted. medical laboratory scientists with a desire to specialize.
Medical laboratory scientists assist physicians with bone
marrow collection and prepare, stain, and microscopically
ENDOTHELIAL CELLS
review bone marrow smears (see Chapter 16). Bone marrow
Because they are structural and do not flow in the bloodstream, aspirates and biopsy specimens are collected and stained to
endothelial cells, the endodermal cells that form the inner analyze nucleated cells that are precursors to blood cells. Cells
surface of the blood vessel, are seldom studied by medical of the erythroid series are precursors to RBCs (see Chapter 8);
laboratory scientists in the hematology laboratory. Neverthe- myeloid series cells mature to form bands and segmented neu-
less, endothelial cells are important in maintaining normal trophils, eosinophils, and basophils (see Chapter 12); and
blood flow, in snaring platelets during times of injury, and in megakaryocytes produce platelets (see Chapter 13). Laboratory
enabling WBCs to escape from the vessel to the surrounding scientists, clinical pathologists, and hematologists review
tissue when called upon. Increasingly refined laboratory Wright-stained aspirate smears for morphologic abnormalities,
methods will enable us to examine fully at least the secretions high or low bone marrow cell concentration, and inappro
(cytokines) of these important cells. priate cell line distributions. An increase in the erythroid
cell line may indicate bone marrow compensation for abnor-
mally increased RBC consumption during blood loss (see
COAGULATION
Chapter 18). The biopsy specimen, enhanced by hematoxylin
Most hematology laboratories provide a blood coagulation and eosin (H&E) staining, may reveal abnormalities in bone
testing department (see Chapters 41, 42, and 45). Plasma marrow architecture indicating leukemia, aplastic anemia, or
coagulation is one component of hemostasis; another is one of a host of other hematologic disorders. Results of exami-
platelets, as reviewed previously. The coagulation system nation of bone marrow aspirates and biopsy specimens are
employs a complex sequence of plasma proteins, some compared with CBC results generated from the peripheral
enzymes, and some enzyme cofactors to produce clot forma- blood to establish commonalities and develop pattern-based
tion after blood vessel injury. Another six to eight enzymes diagnoses.
6 PART I Introduction to Hematology

In the bone marrow laboratory, cytochemical stains may be Chapters 26 and 27). One of the oldest hematology tests, the
employed to differentiate abnormal myeloid, erythroid, and erythrocyte sedimentation rate, detects inflammation and roughly
lymphoid cells (see Chapter 30). These stains include myeloper- estimates its intensity (see Chapter 14).
oxidase, Sudan black B, nonspecific and specific esterase, periodic Finally, the hematology laboratory scientist reviews the cel-
acidSchiff, tartrate-resistant acid phosphatase, and alkaline phos- lular counts, distribution, and morphology in body fluids
phatase. The cytochemical stains are time honored stains and other than blood (see Chapter 17). These include cerebrospinal
are gradually being replaced by flow cytometry immunophe- fluid, synovial (joint) fluid, pericardial fluid, pleural fluid, and
notyping, cytogenetics, and molecular diagnosis (see Chapters peritoneal fluid, in which RBCs and WBCs may be present in
31 to 34). Since 1980, immunostaining methods have enabled disease and in which additional malignant cells may be present
us to identify cell lines, particularly lymphocyte precursors, that require specialized detection skills. Analysis of non-blood
with certainty. An example of immunostaining is an immune- body fluids is always performed with a speedy turnaround,
based dye bound to antibodies to coagulation factor VIII, because cells in these hostile environments rapidly lose their
which is present in megakaryocytes and may be diagnostic for integrity. The conditions leading to a need for body fluid with-
megakaryoblastic leukemia. drawal are invariably acute.
Flow cytometers may be quantitative, such as clinical flow
cytometers that have grown from the original Coulter princi-
HEMATOLOGY QUALITY ASSURANCE
ple, or qualitative, including laser-based instruments that have
AND QUALITY CONTROL
developed from research applications (see Chapter 39). The
former devices are automated clinical profiling instruments Medical laboratory scientists employ particularly complex
that generate the quantitative parameters of the CBC through quality control systems in the hematology laboratory (see
application of electrical impedance and laser or light beam Chapter 5). Owing to the unavailability of weighed standards,
interruption (see Chapter 39). Qualitative laser-based flow the measurement of cells and biologic systems defies chemical
cytometers are mechanically simpler, but technically more standardization and requires elaborate validation, matrix
demanding. Both qualitative and quantitative flow cytometers effect examination, linearity, and reference interval determina-
are employed to analyze cell populations by measuring the tions. An internal standard methodology known as the moving
effects of individual cells on laser light, such as forward-angle average also arose in hematology laboratory applications.6
fluorescent light scatter and right-angle fluorescent light scatter, and Laboratory scientists compare methods through clinical effi-
by immunophenotyping for cell membrane epitopes. The qualita- cacy calculations that produce clinical sensitivity, specificity,
tive flow cytometry laboratory is indispensable to leukemia and positive and negative predictive values for each assay.
and lymphoma diagnosis. Scientists must monitor specimen integrity and test ordering
Molecular diagnostic techniques enhance and even replace patterns and ensure the integrity of reports, including numeri-
some of the advanced hematologic methods. Polymerase chain cal and narrative statements and reference interval compari-
reaction, real-time polymerase chain reaction, microarrays, and sons. As in most branches of laboratory science, the hematology
sequencing systems enable laboratory scientists to detect muta- laboratory places an enormous responsibility for accuracy,
tions such as BCR/ABL fusion, JAK2, and the t(15;17) translo- integrity, judgment, and timeliness on the medical laboratory
cation that signal specific forms of leukemia and establish their professional.
therapeutic profile and prognosis (see Chapters 32 and 34).

REFERENCES
ADDITIONAL HEMATOLOGY PROCEDURES
1. Rhan DH: Examination of the blood. In Lichtman MA,
Laboratory scientists provide several specific manual whole- Beutler E, Kipps TJ, et al, editors: Williams hematology, ed 7,
blood methods to support hematologic diagnosis. The osmotic New York, 2006, McGraw-Hill Medical.
fragility test uses graduated concentrations of saline solutions 2. Wintrobe MM: Hematology, the blossoming of a science,
to detect spherocytes, RBCs with proportionally reduced Philadelphia, 1985, Lea & Febiger.
3. Bizzozero J: ber einem neuen formbestandtheil des blutes
surface membrane area, in hereditary spherocytic or warm autoim- und dessen rolle bei der Thrombose und der Blutgerinnung.
mune hemolytic anemia (see Chapters 23 and 25). Likewise, the Virchows Arch Pathol Anat Physiol Klin Med 90:261-332, 1882.
glucose-6-phosphate dehydrogenase assay tests for an inherited 4. Woronzoff-Dashkoff KK: The Wright-Giemsa stain. Secrets
RBC enzyme deficiency causing severe episodic hemolytic revealed. Clin Lab Med 22:15-23, 2002.
anemia (see Chapter 23). The sickle cell solubility screening 5. Blades AN, Flavell HC: Observations on the use of the Coulter
model D electronic cell counter in clinical haematology. J Clin
assay and its follow-up test, Hb electrophoresis, are used to Pathol 16:158-163, 1963.
detect and diagnose sickle cell anemia and other inherited 6. Gulati GL, Hyun BH: Quality control in hematology. Clin Lab
qualitative hemoglobin abnormalities and thalassemias (see Med 6:675-688, 1986.
Safety in the Hematology Laboratory
Sheila A. Finch*
2
OUTLINE OBJECTIVES
Standard Precautions After completion of this chapter, the reader will be able to:
Applicable Safety
Practices Required by 1. Define standard precautions. 8. Name the most important practice to prevent the
the OSHA Standard 2. List infectious materials included in standard spread of infection.
Housekeeping precautions. 9. Given a written laboratory scenario, assess it for
Laundry 3. Describe the safe practices required in the Occupa- safety hazards and recommend corrective action.
Hepatitis B Virus tional Exposure to Bloodborne Pathogens Standard. 10. Select the proper class of fire extinguisher for a given
Vaccination 4. Identify occupational hazards that exist in the hema- type of fire.
Training and tology laboratory. 11. Define material safety data sheets (MSDSs), list
Documentation 5. Identify the requirements of the Occupational Exposure information contained on MSDSs, and determine
Regulated Medical Waste to Hazardous Chemicals in Laboratories Standard. when MSDSs would be used in laboratory activity.
Management
6. Discuss the development of a safety management 12. Name the specific practice during which most needle
Occupational Hazards
program. stick injuries occur.
Fire Hazard
Chemical Hazards 7. Describe the principles of a fire prevention program,
Electrical Hazard including details such as the frequency of testing
Needle Puncture equipment.
Developing a Safety
Management Program
CASE STUDY
Planning Stage: Hazard
Assessment and After studying the material in this chapter, the reader should be able to respond to the following
Regulatory Review case study:
Safety Program Elements
Hematology Services, Inc., had a proactive safety program. Quarterly safety audits were conducted
by members of the safety committee. The following statements were recorded in the safety
audit report. Which statements describe good work practices, and which statements represent
deficiencies? List the corrective actions that should be taken.
1. A hematology technologist was observed removing gloves and immediately left the laboratory
for a meeting. The medical laboratory professional did not remove the laboratory coat.
2. Food was found in the specimen refrigerator.
3. Syringes were found in the proper sharps container. On further investigation, 50% of the
attached needles were recapped.
4. Hematology technologists were seen in the lunchroom wearing laboratory coats.
5. Fire extinguishers were found every 75 feet of the laboratory.
6. Fire extinguishers were inspected quarterly and annually.
7. One fire drill was conducted in the last 8 months.
8. Unlabeled bottles were found at the workstation.
9. A 1:10 solution of bleach was found near the electronic cell counter. Further investigation
revealed that the bleach solution was made 6 months ago.
10. Gloves were worn by the staff receiving specimens.
11. Material safety data sheets were obtained by fax.
12. Chemicals were stored alphabetically.
13. Fifty percent of the staff interviewed had not participated in a fire drill.

*The author acknowledges the assistance of Debra Walters, safety officer at Indiana University Health, Indianapolis, Ind., for review of this chapter.

7
8 PART I Introduction to Hematology

M
any conditions in the laboratory have the potential 1. Handwashing is one of the most important safety practices.
for causing injury to staff and damage to the build- Hands must be washed with soap and water. If water is
ing or to the community. Patients specimens, not readily available, alcohol hand gels (minimum 62%
needles, chemicals, electrical equipment, reagents, and glass- alcohol) may be used. Hands must be thoroughly dried.
ware all are potential causes of accidents or injury. Managers The proper technique for handwashing is as follows:
and employees must be knowledgeable about safe work a. Wet hands and wrists thoroughly under running
practices and incorporate these practices into the operation of water.
the hematology laboratory. The key to prevention of accidents b. Apply germicidal soap and rub hands vigorously for at
and laboratory-acquired infections is a well-defined safety least 15 seconds, including between the fingers and
program. around and over the fingernails (Figure 2-1, A).
Safety is a broad subject and cannot be covered in one c. Rinse hands thoroughly under running water in a down-
chapter. This chapter simply highlights some of the key safe ward flow from wrist to fingertips (see Figure 2-1, B).
practices that should be followed in the laboratory. Omission d. Dry hands with a paper towel (see Figure 2-1, C). Use
of a safe practice from this chapter does not imply that it is not the paper towel to turn off the faucet handles (see
important or that it should not be considered in the develop- Figure 2-1, D).
ment of a safety curriculum or a safety program. Hands must be washed:
a. Whenever there is visible contamination with blood or
body fluids
STANDARD PRECAUTIONS
b. After completion of work
One of the greatest risks associated with the hematology labo- c. After gloves are removed and between glove changes
ratory is the exposure to blood and body fluids. In December d. Before leaving the laboratory
1991, the Occupational Safety and Health Administration e. Before and after eating and drinking, smoking, apply-
(OSHA) issued the final rule for the Occupational Exposure to ing cosmetics or lip balm, changing a contact lens, and
Bloodborne Pathogens Standard. The rule that specifies stan- using the lavatory
dard precautions to protect laboratory workers and other f. Before and after all other activities that entail hand
healthcare professionals became effective on March 6, 1992. contact with mucous membranes, eyes, or breaks in
Universal precautions was the original term; OSHAs current skin
terminology is standard precautions. Throughout this text, the 2. Eating, drinking, smoking, and applying cosmetics or lip balm
term standard precautions is used to remind the reader that all must be prohibited in the laboratory work area.
blood, body fluids, and unfixed tissues are to be handled as 3. Hands, pens, and other fomites must be kept away from
though they were potentially infectious. the workers mouth and all mucous membranes.
Standard precautions must be adopted by the laboratory. 4. Food and drink, including oral medications and tolerance-
Standard precautions apply to the following potentially infec- testing beverages, must not be kept in the same refrigerator
tious materials: blood, semen, vaginal secretions, cerebrospinal as laboratory specimens or reagents or where potentially
fluid, synovial fluid, pleural fluid, any body fluid with visible infectious materials are stored or tested.
blood, any unidentified body fluid, unfixed slides, microhema- 5. Mouth pipetting must be prohibited.
tocrit clay, and saliva from dental procedures. Past practice was 6. Needles and other sharp objects contaminated with blood
to label specimens from patients known to have infectious and other potentially infectious materials should not be
diseases; however, experience has shown that patients without manipulated in any way. Such manipulation includes
visible symptoms can harbor infectious diseases. Labeling such resheathing, bending, clipping, or removing the sharp
specimens also jeopardizes patient confidentiality. Adopting object. Resheathing or recapping is permitted only when
standard precautions lessens the risk of healthcare worker there are no other alternatives or when the recapping is
exposures to blood and body fluids, decreasing the risk of required by specific medical procedures. Recapping is
injury and illness. permitted by use of a method other than the traditional
Bloodborne pathogens are pathogenic microorganisms two-handed procedure. The one-handed method or a
that, when present in human blood, can cause disease. They resheathing device is often used (see Chapter 3). Docu-
include, but are not limited to, hepatitis B virus (HBV), hepa- mentation in the exposure control plan should identify the
titis C virus, and human immunodeficiency virus (HIV). This specific procedure by which resheathing is permitted.
chapter does not cover the complete details of the standard; it 7. Contaminated sharps (including, but not limited to,
discusses only the sections that apply directly to the hemato needles, blades, pipettes, syringes with needles, and
logy laboratory. Additional information can be found in the glass slides) must be placed in a puncture-resistant con-
references at the end of this chapter. tainer that is appropriately labeled with the universal
biohazard symbol (Figure 2-2) or a red container that
Applicable Safety Practices Required adheres to the standard. The container must be leak-proof
by the OSHA Standard (Figure 2-3).
The following standards are applicable in a hematology labora- 8. Procedures such as removing caps when checking for clots,
tory and must be enforced: filling hemacytometer chambers, making slides, discarding
CHAPTER 2 Safety in the Hematology Laboratory 9

A B

C D
Figure 2-1 Proper handwashing technique. A, Wet hands thoroughly under running water, apply soap, and rub hands vigorously for at least 15 seconds.
B, Rinse hands thoroughly under running water in a downward flow from wrist to fingertips. C, Dry hands with a paper towel. D, Turn off faucet with paper
towel. (From Young AP, Proctor DB: Kinns the medical assistant, ed 11, St Louis, 2011, Saunders.)

specimens, making dilutions, and pouring specimens or


fluids must be performed so that splashing, spraying, or
production of droplets of the specimen being manipulated
is prevented. These procedures may be performed behind
a barrier, such as a plastic shield, or protective eyewear
should be worn (Figure 2-4).
9. Personal protective clothing and equipment must be pro-
vided to the worker. The most common forms of personal
protective equipment are described in the following:
a. Outer coverings, including gowns, laboratory coats, and
sleeve protectors, should be worn when there is a
chance of splashing or spilling on work clothing. The
outer covering must be made of fluid-resistant material,
must be long-sleeved, and must remain buttoned at all
times. If contamination occurs, the personal protective
equipment should be removed immediately and treated
as infectious material.
Cloth laboratory coats may be worn if they are fluid
resistant. If cloth coats are worn, the coats must be
laundered inside the laboratory or hospital or by a
contracted laundry service. Laboratory coats used in the
laboratory while performing laboratory analysis are
considered personal protective equipment and are not
Figure 2-2 Biohazard symbol. to be taken home.
10 PART I Introduction to Hematology

A B

C D
Figure 2-3 Examples of sharps disposal systems. A, Molded foot pedal cart with hinged or slide top lid. B, In-Room system wall enclosures. C, Multipurpose
container with horizontal drop lid. D, Phlebotomy containers. (Courtesy Covidien, Mansfield, Mass.)

All protective clothing should be removed before hand between the glove and the hand and slipping the
the worker leaves the laboratory; it should not be second glove off. This technique prevents contamina-
worn into public areas. Public areas include, but are tion of the clean hand by the dirty second glove
not limited to, break rooms, storage areas, bathrooms, (Figure 2-5).1 Contaminated gloves should be disposed
cafeterias, offices, and meeting places outside the of according to applicable federal or state regulations.
laboratory. c. Eyewear, including face shields, goggles, and masks,
A second laboratory coat can be made available for should be used when there is potential for aerosol
use in public areas. A common practice is to have a mists, splashes, or sprays to mucous membranes
different-colored laboratory coat that can be worn in (mouth, eyes, or nose). Removing caps from specimen
public areas. This second laboratory coat would be tubes, working at the cell counter, and centrifuging
laundered by the employee. specimens are examples of tasks that could result in
b. Gloves must be worn when the potential for contact creation of aerosol mist.
with blood or body fluids exists (including when 10. Phlebotomy trays should be appropriately labeled to indi-
removing and handling bagged biohazardous material cate potentially infectious materials. Specimens should be
and when decontaminating bench tops) and when placed into a secondary container, such as a resealable
venipuncture or finger sticks are performed. One of the biohazard-labeled bag.
limitations of gloves is that they do not prevent needle 11. If a pneumatic tube system is used to transport specimens,
sticks or other puncture wounds. Provision of gloves to the specimens should be transported in the appropriate
laboratory workers must accommodate latex allergies. tube (primary containment), placed into a special self-
Alternative gloves must be readily accessible to any sealing leak-proof bag, appropriately labeled with the bio-
laboratory worker with a latex allergy. Gloves must be hazard symbol (secondary containment). Requisition
changed after each contact with a patient, when there forms should be placed outside of the secondary container
is visible contamination, and when physical damage to prevent contamination if the specimen leaks. Foam
occurs. Gloves should not be worn when clean inserts for the pneumatic tube system carrier prevent shift-
devices, such as a copy machine or a clean telephone, ing of the sample during transport and also act as a shock
are used. Gloves must not be worn again or washed. absorber, thus decreasing the risk of breakage.
After one glove is removed, the second glove can be When specimens are received in the laboratory,
removed by sliding the index finger of the ungloved they should be handled by someone wearing gloves, a
CHAPTER 2 Safety in the Hematology Laboratory 11

bleach, used in a 1:10 volume/volume dilution (10%), which


can be made by adding 10mL of bleach to 90mL of water or
2 1 2 cups of bleach to 1 gallon of water to achieve the recom-
mended concentration of chlorine of 5500ppm. Because this
solution is not stable, it must be made fresh daily. The con-
tainer of 1:10 solution of bleach should be labeled properly
with the name of the solution, the date and time prepared, the
date and time of expiration (24 hours), and the initials of the
preparer. Bleach is not recommended for aluminum surfaces.
Other solutions used to decontaminate include, but are not
limited to a phenol-based disinfectant such as Amphyl, tuber-
culocidal disinfectants, and 70% ethanol. All paper towels used
in the decontamination process should be disposed of as bio-
A
hazardous waste. Documentation of the disinfection of work
areas and equipment after each shift is required.

Laundry
If nondisposable laboratory coats are used, they must be placed
in appropriate containers for transport to the laundry at the
facility or to a contract service and not taken to the employees
home.

Hepatitis B Virus Vaccination


Laboratory workers should receive the HBV vaccination series
at no cost before or within 10 days after beginning work in the
laboratory. An employee must sign a release form if he or she
B refuses the series. The employee can request and receive the
hepatitis vaccination series at any time, however. If an exposure
Figure 2-4 Examples of safety shields. A, Face shield. B, Adjustable incident (needle puncture or exposure to skin, eye, face, or
swing arm shield. (Courtesy Steve Kasper.) mucous membrane) occurs, postexposure evaluation and
follow-up, including prophylaxis and medical consultation,
should be made available at no cost to the employee. Employ-
laboratory coat, and other protective clothing, in accor- ees should be encouraged to report all exposure incidents, and
dance with the type and condition of specimen. Contami- such reporting should be enforced as standard policy.
nated containers or requisitions must be decontaminated
or replaced before being sent to the work area. Training and Documentation
12. When equipment used to process specimens becomes Hematology staff should be properly educated in epidemio
visibly contaminated or requires maintenance or service, logy and symptoms of bloodborne diseases, modes of trans-
it must be decontaminated, whether service is performed mission of bloodborne diseases, use of protective equipment,
within the laboratory or by a manufacturer repair service. work practices, ways to recognize tasks and other activities that
Decontamination of equipment would consist at a may result in an exposure, and the location of the written
minimum of flushing the lines and wiping the exterior and exposure plan for the laboratory. Education should be docu-
interior of the equipment. If it is difficult to decontaminate mented and should occur when new methods, equipment, or
the equipment, it must be labeled with the biohazard procedures are introduced; at the time of initial assignment to
symbol to indicate potentially infectious material. Routine the laboratory; and at least annually thereafter.
cleaning should be performed on equipment that has the
potential for receiving splashes or sprays, such as inside Regulated Medical Waste Management
the lid of the microhematocrit centrifuge. Specimens from the hematology laboratory are identified as
regulated waste. There are different categories of regulated
Housekeeping medical waste, and state and local regulations for disposal of
Blood and other potentially infectious materials can contami- medical waste must be followed. OSHA regulates some aspects
nate work surfaces easily. Contamination can be caused by of regulated medical waste such as needle handling, occupa-
splashes, poor work practices, and droplets of blood on the tional exposure, labeling of containers, employee training, and
work surface. To prevent contamination, all work surfaces storing of the waste. The Occupational Exposure to Blood-
should be cleaned when procedures are completed and borne Pathogens Standard provides information on the
whenever the bench area or floor becomes visibly contami- handling of regulated medical waste. Specific disposal guide-
nated. An appropriate disinfectant solution is household lines are specific to the state disposal standards. When two
12 PART I Introduction to Hematology

A B

C D
Figure 2-5 Removal of gloves. A, Using one hand, grasp the outside of the other glove and slowly pull it off the hand, turning it inside out as you remove
it. B, Scrunch the removed glove into a ball. C, Place the index and middle finger of the ungloved hand on the inside of the other glove. D, Pull the second
glove off of the hand, turning it inside out as it is removed and enclosing the balled-up glove. (From Bonewit-West K: Clinical procedures for medical assistants,
ed 7, St Louis, 2008, Saunders.)

regulations conflict it is recommended that the more stringent TABLE 2-1 Fire Extinguisher Classifications
standard be followed.
Class/Type of
Extinguisher Type of Fire
OCCUPATIONAL HAZARDS
A Use class A extinguishers on fires involving ordinary
Four important occupational hazards in the laboratory are dis- combustibles such as wood, cloth, or paper.
cussed in this chapter: fire hazard, chemical hazards, electrical B Use class B extinguishers on fires involving
hazard, and needle puncture. There are other hazards to be flammable liquids, gases, or grease.
considered when a safety management program is developed, C Use class C extinguishers on energized (plugged-in)
and the reader is referred to the Department of Labor section electrical fires. Examples are fires involving
of the Code of Federal Regulations for detailed regulations.2 household appliances, computer equipment, fuse
boxes, or circuit breakers.
Fire Hazard ABC ABC extinguishers are multipurpose extinguishers
Because of the numerous flammable and combustible chemi- that handle type A, B, and C fires.
cals used in the laboratory, fire is a potential hazard. Comply-
ing with standards established by the National Fire Protection
Association, OSHA, the Joint Commission, the College of should be checked monthly and maintained annually. Not
American Pathologists, and other organizations can minimize all fire extinguishers are alike. Each fire extinguisher is rated
the dangers. A good fire safety/prevention plan is necessary and for the type of fire that it can suppress. It is important to
should consist of the following: use the correct fire extinguisher for the given class of fire.
1. Enforcement of a no-smoking policy. Hematology laboratory workers should be trained to recog-
2. Installation of appropriate fire extinguishers. Several types nize the class of extinguisher and to use a fire extinguisher
of extinguishers, most of which are multipurpose, are avail- properly. Table 2-1 summarizes the fire extinguisher clas-
able for use for specific types of fire. sifications. The fire extinguishers used in the laboratory are
3. Placement of fire extinguishers every 75 feet. A distinct portable extinguishers and are not designed to fight large
system for marking the locations of fire extinguishers fires. In the event of a fire in the laboratory, the local fire
enables quick access when they are needed. Fire extinguishers department must be contacted immediately.
CHAPTER 2 Safety in the Hematology Laboratory 13

4. Placement of adequate fire detection systems (alarms, sprin- chemical, preparation or fill date, expiration date (time, if
klers), which should be tested every 3 months. applicable), initials of preparer (if done in-house), and
5. Placement of manual fire alarm boxes near the exit doors. chemical hazards (e.g., poisonous, corrosive, flammable).
Travel distance should not exceed 200 feet. Do not use a chemical that is not properly labeled as to
6. Written fire prevention and response procedures, com- identity or content.
monly referred to as the fire response plan. All staff in the 2. Follow all handling and storage requirements for the
laboratory should be knowledgeable about the procedures. chemical.
Workers should be given assignments for specific responsi- 3. Store alcohol and other flammable chemicals in approved
bilities in case of fire, including responsibilities for patient safety cans or storage cabinets at least 5 feet away from a
care, if applicable. Total count of employees in the labora- heat source (e.g., Bunsen burners, paraffin baths). Limit
tory should be known for any given day, and a buddy the quantity of flammable chemicals stored on the work-
system should be developed in case evacuation is necessary. bench to 2 working days supply. Do not store chemicals
Equipment shutdown procedures should be addressed in in a hood or in any area where they could react with other
the plan, as should responsibility for implementation of chemicals.
those procedures. 4. Use adequate ventilation, such as fume hoods, when
7. Fire drills, which should be conducted so that response to working with hazardous chemicals.
a fire situation is routine and not a panic response. Fre- 5. Use personal protective equipment, such as gloves (e.g.,
quency of fire drills varies by type of occupancy of the nitrile, polyvinyl chloride, rubberas appropriate for
building and by accrediting agency. Overall governance is the chemical in use), rubber aprons, and face shields.
by the state fire marshall. All laboratory staff members Safety showers and eye wash stations should be available
should participate in the fire drills. Proper documentation every 100 feet or within 10 seconds of travel distance
should be maintained to show that all phases of the fire from every work area where the hazardous chemicals are
response plan were activated. If patients are in the hemato used.
logy laboratory, evacuation can be simulated, rather than 6. Use bottle carriers for glass bottles containing more than
evacuating actual patients. The entire evacuation route 500mL of hazardous chemical.
should be walked to verify the exit routes and clearance of 7. Use alcohol-based solvents, rather than xylene or other
the corridors. A summary of the laboratorys fire response particularly hazardous substances, to clean microscope
plan can be copied onto a quick reference card and attached objectives.
to workers identification badges to be readily available in 8. The wearing of contact lenses should not be permitted
a fire situation. when an employee is working with xylene, acetone, alco-
8. Written storage requirements for any flammable or com hols, formaldehyde, and other solvents. Many lenses are
bustible chemicals stored in the laboratory. Chemicals permeable to chemical fumes. Contact lenses can make it
should be arranged according to hazard class and not difficult to wash the eyes adequately in the event of a
alphabetically. splash.
9. A well-organized fire safety training program. This 9. Spill response procedures should be included in the
program should be completed by all employees. Activities chemical safety procedures, and all employees must receive
that require walking evacuation routes and locating fire training in these procedures. Absorbent material should be
extinguishers and pull boxes in the laboratory area should available for spill response. Multiple spill response kits
be scheduled. Types of fires likely to occur and use of and absorbent material should be stored in various areas
the fire extinguisher should be discussed. Local fire and rooms rather than only in the area where they are
departments work with facilities to conduct fire safety likely to be needed. This prevents the need to walk through
programs. the spilled chemical to obtain the kit.
10. Material safety data sheets (MSDSs) are written by the
manufacturers of chemicals to provide information on the
Chemical Hazards chemicals that cannot be put on a label. When an MSDS
Some of the chemicals used in the hematology laboratory are is received in the laboratory, it must be retained and
considered hazardous and are governed by the Occupational reviewed with laboratory personnel. The MSDS provides
Exposure to Hazardous Chemicals in Laboratories Standard. information on the following:
This standard requires laboratories to develop a chemical a. Manufacturername, address, emergency phone
hygiene plan that outlines safe work practices to minimize number, date prepared
exposures to hazardous chemicals. The full text of this standard b. Hazardous ingredientscommon names, worker expo-
can be found in 29 CFR (Code of Federal Regulations) sure limits
1910.1450. c. Physical and chemical characteristicsboiling point,
General principles that should be followed in working with vapor pressure, evaporation rate, appearance, and odor
chemicals are as follows: under normal conditions
1. Label all chemicals properly, including chemicals in sec- d. Physical hazardsdata on fire and explosion hazard
ondary containers, with the name and concentration of the and ways to handle the hazards
14 PART I Introduction to Hematology

e. Reactivitystability of the chemical and with what 8. Equipment that causes shock or a tingling sensation should
chemicals it will react be turned off, the instrument unplugged and identified as
f. Health hazardssigns and symptoms of exposure, defective, and the problem reported.
such as eye irritation, nausea, dizziness, and headache 9. Before repair or adjustment of electrical equipment is
g. Documentation on the MSDS of whether the reagent attempted, the following should be done:
or a component of the reagent is considered a carcino- a. Unplug the equipment.
gen, teratogen, or mutagen, so that process and proce- b. Make sure the hands are dry.
dures are adopted to limit risk of exposure c. Remove jewelry.
h. Precautions for safe handling and usewhat to do if
the chemical spills, how to dispose of the chemical, and Needle Puncture
what equipment is required for cleanup Needle puncture is a serious occupational hazard for labora-
i. Control measureshow to reduce the exposure to tory workers. Needle-handling procedures should be written
the chemical and protective clothing or equipment, and followed, with special attention to phlebotomy procedures
such as respirator, gloves, and eye protection, that and disposal of contaminated needles (see Chapter 3). Other
should be used in handling the chemical. See items that can cause a puncture similar to a needle puncture
Appendix A for a sample MSDS. Use this sample to include sedimentation rate tubes, applicator sticks, capillary
review the information that can be found on the tubes, glass slides, and transfer pipettes.
MSDS. Disposal procedures should be followed and enforced. The
An MSDS management system should be consid- most frequent cause of a needle puncture or a puncture from
ered to track the incoming MSDSs received in the labo- other sharp objects is improper disposal. Failure to check
ratory. When new or revised MSDSs are received, a sharps containers on a regular basis and to replace them when
notice should be posted to alert the hematology staff they are no more than three quarters full encourages overstuff-
that new or revised MSDSs have been received. MSDSs ing, which sometimes leads to injury. Portable bedside con-
may be obtained electronically by means of computer, tainers are available for workers when performing venipunctures
fax, Internet, or CD-ROM. If an electronic device is used or capillary punctures. Wall-mounted needle disposal con
in the laboratory to receive and store MSDSs, each tainers also are available and make disposal convenient. As
employee must be trained on the use of the device. The mentioned previously, all needle punctures should be reported
training must include emergency procedures in case of to the health services or proper authorities within the
power outages or malfunctions of the device. The institution.
device must be reliable and readily accessible during
the hours of operation. In the event of emergency, hard
copies of the MSDSs must be accessible to medical staff.
DEVELOPING A SAFETY
MSDSs are required to be kept for 30 years after employ-
MANAGEMENT PROGRAM
ment of the last employee who used the chemicals in
the work area, and they should be documented with Every accredited laboratory is required to have a safety manage-
the date when the chemical is no longer used in the ment program. A safety management program is one that
laboratory. identifies the guidelines necessary to provide a safe working
environment free from recognizable hazards that can cause
harm or injury. Many medical laboratory scientists assume
Electrical Hazard positions as supervisors or laboratory safety officers. Responsi-
Electrical equipment and outlets are other sources of hazard. bilities in these positions require knowledge of the safety prin-
Faulty wiring may cause fires or serious injury. Guidelines ciples and the development, implementation, and maintenance
include the following: of a laboratory safety program. This section provides an over-
1. Equipment must be grounded or double insulated. view of the elements that should be considered in developing
(Grounded equipment has a three-pronged plug.) a safety program.
2. Use of cheater adapters (adapters that allow three-
pronged plugs to fit into a two-pronged outlet) should be Planning Stage: Hazard Assessment and
prohibited. Regulatory Review
3. Use of gang plugs (plugs that allow several cords to be Assessment of the hazards found in the laboratory and aware-
plugged into one outlet) should be prohibited. ness of the standards and regulations that govern laboratories
4. Use of extension cords should be avoided. is a required step in the development of a safety program.
5. Equipment with loose plugs or frayed cords should not be Taking the time to become knowledgeable about the regula-
used. tions and standards that relate to the procedures performed in
6. Stepping on cords, rolling heavy equipment over cords, and the hematology laboratory is an essential first step. Examples
other abuse of cords should be prohibited. of the regulatory agencies that have standards, requirements,
7. When cords are unplugged, the plug, not the cord, should and guidelines that are applicable to hematology laboratories
be pulled. are given in Box 2-1. Sorting through the regulatory maze
CHAPTER 2 Safety in the Hematology Laboratory 15

environmental hazards that exist in the hematology


BOX 2-1 Government Regulatory Agencies Providing laboratory.
Laboratory Safety Standards Training programsconducted annually for all employees.
New employees should receive safety information on
Department of Labor: 29 Code of Federal the first day that they are assigned to the hematology
Regulations Parts 1900-1910 laboratory.
Hazard Communication Standard (right to know)29 CFR 1910.1200 Job safety analysisidentifies all of the tasks performed in the
Occupational Exposure to Bloodborne Pathogens Standard29 CFR hematology laboratory, the steps involved in performing the
1910.1030 procedures, and the risk associated with the procedures.
Occupational Exposure to Hazardous Chemicals in Laboratories Safety awareness programpromotes a team approach and
Standard29 CFR 1910.1450 encourages employees to take an active part in the safety
Formaldehyde Standard29 CFR 1910.1048 program.
Air Contaminants: Permissible Exposure Limits29 CFR 1910.1000 Risk assessmentproactive risk (identification) assessment
Occupational Noise Level Standard29 CFR 1910.95 of all the potential safety, occupational, or environmental
Hazardous Waste Operations and Emergency Response Standard29 hazards that exist in the laboratory. The assessment should
CFR 1910.120 be conducted at least annually and when a new risk is added
Personal Protective Equipment29 CFR 1910.132 to the laboratory. After the risk assessment is conducted,
Eye and Face Protection29 CFR 1910.133 goals, policies, and procedures should be developed to
Respiratory Protection29 CFR 1910.134 prevent the hazard from injuring a laboratory worker. Some
common risks are exposure to bloodborne pathogens; expo-
Medical Waste Standards Regulated sure to chemicals; needle punctures; slips, trips, and falls;
by the State and ergonomics issues.
State medical waste standards Safety audits and follow-upa safety checklist should be
developed for the hematology laboratory.
Department of the Interior, Environmental Reporting and investigating of all accidents, near misses, or
Protection Agency: 40 Code of Federal unsafe conditionsthe causes of all incidents should be
Regulations Parts 200-399 reviewed and corrective action taken, if necessary.
Resource Conservation and Recovery Act (RCRA) Emergency drill and evaluationperiodic drills for all poten-
Clean Air Act tial internal and external disasters. Drills should address the
Clean Water Act potential accident or disaster before it occurs and test the
Toxic Substances Control Act (TSCA) preparedness of the hematology workers for an emergency
Comprehensive Environmental Response, Compensation, and Liability situation. Planning for the accident and practicing the
Act (CERCLA) response to the accident reduces the panic that results when
Superfund Amendments and Reauthorization Act (SARA) the correct response is not followed.
SARA Title III: Community Right to Know Act Emergency management planemergencies, sometimes
called disasters (anything that prevents normal operation of
Voluntary Agencies/Accrediting Agencies/Other the laboratory), do not occur only in the hospital-based
Government Agencies laboratories. Freestanding laboratories, physician office
The Joint Commission laboratories, and university laboratories can be affected by
College of American Pathologists (CAP) emergencies that occur in the building or in the community.
State public health departments Emergency planning is crucial to being able to experience
Centers for Disease Control and Prevention (CDC) an emergency situation and recover enough to continue the
Clinical and Laboratory Standards Institute daily operation of the laboratory. In addition to the safety
National Fire Protection Association (NFPA) risk assessment, a hazard vulnerability analysis should be
Department of Transportation (DOT): Regulated Medical Waste Shipment conducted. Hazard vulnerability analysis helps to identify
Regulations49 CFR 100-185 all of the potential emergencies that may have an impact
CFR, Code of Federal Regulations. on the laboratory. Emergencies such as a utility failureloss
of power, water, or telephonescan have a great impact on
the laboratorys ability to perform procedures. Emergencies
can be frustrating, but the government agencies and voluntary in the community, such as a terrorist attack, plane crash,
standards organizations are willing to assist employers in com- severe weather, flood, or civil disturbances, can affect the
plying with their standards. laboratory workers ability to get to work and can affect
transportation of crucial supplies or equipment. When the
Safety Program Elements potential emergencies are identified, policies and proce-
A proactive program should include the following elements: dures should be developed and practiced so that the labora-
Written safety planwritten policies and procedures that tory worker knows the backup procedures and can
explain the steps to be taken for all of the occupational and implement them quickly during an emergency or disaster
16 PART I Introduction to Hematology

situation. The emergency management plan should cover


the four phases of response to an emergency, as follows: BOX 2-2 Emergency Management Activities:
1. Mitigationmeasures to reduce the adverse effects of the Planning for Response to a Fire
emergency
Mitigation Tools
2. Preparednessdesign of procedures, identification of
Fire alarm pull box
resources that may be used, and training in the
Emergency code to notify workers
procedures
Smoke detectors
3. Responseactions that will be taken when responding
Fire/smoke doors
to the emergency
Audible and visual alarms
4. Recoveryprocedures to assess damage, evaluate
Fire exit lights
response, and replenish supplies so that the laboratory
Sprinkler system
can return to normal operation
An example of an emergency management plan is Preparedness Activities
shown in Box 2-2. The Clinical and Laboratory Standards Training of workers
Institute document Planning for Challenges to Clinical Fire drills
Laboratory Operations During a Disaster describes detailed Fire response procedure development
actions to prepare for an emergency or disaster. The Annual and monthly fire extinguisher checks
document also lists some valuable websites for additional
resources.3 Response Activities
Safety committee/department safety meetingsto communi- Fire response plan implementation
cate safety policies to the employees. Assignment of specific tasks during the actual event
Review of equipment and supplies purchased for the laboratory
for code compliance and safety features. Recovery Activities
Annual evaluation of the safety programreview of goals and Communication of all clear
performance as well as a review of the regulations to assess Documentation of response to the fire
compliance in the laboratory. Damage assessment
Financial accounting of response activities
Replenishment of supplies
Stress debriefing for workers

SUMMARY
The responsibility of a medical laboratory professional is to perform Clean up spills immediately, if substance is low hazard and
analytic procedures accurately, precisely, and safely. spill is small; otherwise contact hazardous materials team
Safe practices must be incorporated into all laboratory procedures (internal or vendor) for spill reporting and appropriate spill
and should be followed by every employee. management.
The laboratory must adopt standard precautions, which require Keep workstations clean and corridors free from obstruction.
that all human blood, body fluids, and unfixed tissues be treated Report injuries and unsafe conditions. Review accidents and
as if they were infectious. incidents to determine their fundamental cause. Take correc-
One of the most important safety practices is handwashing. tive action to prevent further injuries.
Occupational hazards in the laboratory include fire, chemical, and Maintain a proactive safety management program.
electrical hazards and needle puncture.
Some common-sense rules of safety are as follows: Now that you have completed this chapter, go back and
Be knowledgeable about the procedures being performed. If in read again the case study at the beginning and respond
doubt, ask for further instructions. to the questions presented.
Wear protective clothing and use protective equipment when
required.

R E V I E W Q UESTIONS
1. Standard precautions apply to all of the following except: 2. The most important practice in preventing the spread of
a. Blood disease is:
b. Cerebrospinal fluid a. Wearing masks during patient contact
c. Semen b. Proper handwashing
d. Concentrated acids c. Wearing disposable laboratory coats
d. Identifying specimens from known or suspected
HIV- and HBV-infected patients with a red label
CHAPTER 2 Safety in the Hematology Laboratory 17

3. The appropriate dilution of bleach to be used in laboratory 8. It is a busy evening in the City Hospital hematology
disinfection is: department. One staff member called in sick, and there
a. 1:2 was a major auto accident that has one staff member tied
b. 1:5 up in the blood bank all evening. Mary, the medical labo-
c. 1:10 ratory scientist covering hematology, is in a hurry to get a
d. 1:100 stat sample on the analyzer but needs to pour off an
aliquot for another department. She is wearing gloves and
4. How frequently should fire alarms and sprinkler systems a gown. She carefully covers the stopper of the well-mixed
be tested? ethylenediaminetetraacetic acid (EDTA) tube with a gauze
a. Weekly square and tilts the stopper toward her so it opens away
b. Monthly from her. She pours off about 1mL into a prelabeled tube,
c. Quarterly replaces the stopper of the EDTA tube, and puts it in the
d. Annually sample rack and sets it on the conveyor. She then runs the
poured sample off to the other department. How would
5. Where should alcohol and other flammable chemicals be you assess Marys safety practice?
stored? a. Mary was careful and followed all appropriate
a. In an approved safety can or storage cabinet away procedures.
from heat sources b. Mary should have used a shield when opening the
b. Under a hood and arranged alphabetically for ease of tube.
identification in an emergency c. Mary should have poured the sample into a sterile
c. In a refrigerator at 2 C to 8 C to reduce volatilization tube.
d. On a low shelf in an area protected from light d. Mary should have wiped the tube with alcohol after
replacing the stopper.
6. The most frequent cause of needle punctures is:
a. Patient movement during venipuncture 9. A class C fire extinguisher would be appropriate to use on
b. Improper disposal of phlebotomy equipment a fire in a chemical cabinet.
c. Inattention during removal of needle after venipuncture a. True
d. Failure to attach needle firmly to syringe or tube holder b. False

7. Under which of the following circumstances would an 10. According to OSHA standards, laboratory coats must be
MSDS be helpful? all of the following except:
a. A phlebotomist has experienced a needle puncture a. Water resistant
with a clean needle. b. Made of cloth fabric that can be readily laundered
b. A fire extinguisher failed during routine testing. c. Long-sleeved
c. A pregnant laboratory staff member has asked d. Worn fully buttoned
whether she needs to be concerned about working
with a given reagent.
d. During a safety inspection, an aged microscope power
supply is found to have a frayed power cord.

REFERENCES
1. Garza D, Becan-McBride K: Phlebotomy handbook, ed 7, Upper 3. Hearn TL, Astles JR, Kaplan LA, et al: Planning for challenges
Saddle River, NJ, 2005, Pearson Prentice Hall. to clinical laboratory operations during a disaster, a report.
2. Department of Labor: 29 Code of Federal Regulations Parts X4- R, vol 23, no 29. Available at: http://www.clsi.org/source/
1900-1910. Federal Register, July 1, 1998. Available at: http:// orders/free/x04rf.pdf. Accessed June 4, 2010.
www.access.gpo.gov/nara/cfr/waisidx_98/29cfrv5__98.html.
Accessed June 5, 2010.

ADDITIONAL RESOURCES
The Joint Commission Manual: Environment of Care Standards, Resource for four phases of emergency planning and up-to-date
2009. disaster information. Available at: http://www.fema.gov.
National Fire Protection Association: Laboratories Using Chemi- Resource for regulatory health guidelines. Available at: http://
cals, NFPA 45. www.osha.gov/SLTC/healthguidelines.
Resource for occupational hazards found in the laboratory and National Institute for Occupational Safety and Health pocket guide
other related regulations. Available at: http://www.osha.gov/ for chemical hazards. Available at: http://www.cdc.gov/niosh/
SLTC/etools/hospital/lab/lab.html. npg.
3 Specimen Collection*
Carole A. Mullins

OUTLINE OBJECTIVES
Safety After completion of this chapter, the reader will be able to:
Responsibility of the
Phlebotomist in 1. Describe the application of standard precautions to 7. Explain appropriate use of skin puncture equipment
Infection Control the collection of blood samples. and procedure to be followed, including venipuncture
Physiologic Factors 2. List collection equipment used for venipuncture and sites for infants, children, and adults.
Affecting Test Results skin puncture. 8. Discuss essentials of quality assurance in specimen
Venipuncture 3. Correlate tube stopper color with additive, if any, and collection.
Collection Equipment for explain the purpose of the additive and use of that 9. List reasons for specimen rejection.
Venipuncture tube type in the laboratory. 10. Given the description of a specimen and its collec-
Routine Venipuncture 4. Discuss selection of a vein for venipuncture tion, determine specimen acceptability.
Procedure and name the vein that is preferred in most 11. Recognize deviations from the recommended veni-
Venipuncture Procedure
instances. puncture practice in a written scenario and describe
in Children and Infants
5. List in order the steps recommended by the Clinical corrective procedures.
Complications
Encountered in Blood and Laboratory Standards Institute for venipuncture 12. Name the most important step in the venipuncture
Collection in adults, including recommended order of collection procedure.
Skin Punctures for tubes with various additives. 13. List reasons for inability to obtain a blood specimen.
Collection Sites 6. Discuss complications encountered in blood collec- 14. Summarize legal issues that need to be considered
Skin Puncture Technique tion and proper response of the phlebotomist. in specimen acquisition.
Devices for Collecting
Blood by Skin Puncture
Skin Puncture Procedure CASE STUDIES
Preparation of Blood After studying the material in this chapter, the reader should be able to respond to the following
Smears case studies:
Quality Assurance in
Specimen Collection Case 1
Technical Competence A phlebotomist asks an outpatient, Are you Susan Jones? After the patient answers yes, the phle-
Collection Procedures botomist proceeds by labeling the tubes and drawing the blood. What is wrong with this
Anticoagulants and scenario?
Preservatives
Requirements for a Case 2
Quality Specimen
A patient must have blood drawn for a complete blood count (CBC), potassium (K+) level, pro-
Blood Collection Attempts
thrombin time (PT), and type and screen. The phlebotomist draws blood into the following tubes
Collection of Specimens
for Blood Culture in this order:
Quality Control and 1. SST or serum separation tube
Preventive Maintenance 2. Light blue tube for PT
on Specimen Collection 3. Lavender tube for CBC
Instruments 4. Green tube for K+
Reasons for Specimen Which of the results will be affected by the incorrect order of draw? Explain.
Rejection
Specimen Handling
Legal Issues in
Phlebotomy

*The editors thank Dennis J. Ernst, MT(ASCP), director, Center for Phlebotomy Education, Inc., for his review of this chapter.

18
CHAPTER 3 Specimen Collection 19

SAFETY BOX 3-1 Physiologic Factors Affecting Test Results


Standard precautions must be followed in the collection of
blood, and all specimens must be treated as potentially Posture
infectious for bloodborne pathogens (e.g., hepatitis B virus, Changing from a supine (lying) to a sitting or standing position results
hepatitis C virus, and human immunodeficiency virus [HIV]). in a shift of body water from inside the blood vessels to the interstitial
Regulations of the Occupational Health and Safety Administra- spaces. Larger molecules cannot filter into the tissues and concen-
tion (OSHA) that took effect on March 6, 1992, outlined in trate in the blood. There are significant increases in test values for
detail what must be done to protect healthcare workers from lipids, enzymes, and proteins.
exposure to bloodborne pathogens, such as the pathogens that
cause hepatitis C, hepatitis B, hepatitis D, syphilis, malaria, and Diurnal Rhythm
HIV infection.1 Diurnal pertains to daylight, and diurnal rhythm refers to daily body
Bloodborne pathogens may enter the body via an accidental fluid fluctuations that occur. Levels of certain hormones, such as
injury by a sharp object, such as a contaminated needle, a cortisol and adrenocorticotropic hormone, decrease in the afternoon.
scalpel, broken glass, or anything else that can pierce the skin. Other test values, such as iron and eosinophil levels, increase in the
Cuts, skin areas with dermatitis, abrasions, and mucous mem- afternoon.
branes of the mouth, eyes, and nose may provide a portal of
entry. Indirect transmission can occur when a person touches Exercise
a contaminated surface or object and then touches the mouth, Muscle activity elevates creatinine, protein, creatine kinase, aspar-
eyes, nose, or nonintact skin without washing the hands. Hepa- tate transaminase, and lactate dehydrogenase test values. Research
titis B virus can survive on inanimate or dried surfaces at room also suggests that exercise activates coagulation and fibrinolysis and
temperature for at least 1 week.2 increases platelet and white blood cell counts.3
Handwashing is the most important practice to prevent the
spread of infectious diseases. The phlebotomist should wash Stress
his or her hands with a nonabrasive soap and running water Anxiety can cause a temporary increase in white blood cells.2
between patients and every time gloves are removed. If hand-
washing facilities are unavailable, an antiseptic hand cleanser Diet
or an antiseptic towelette may be used as a temporary measure. Fasting means no food or beverages except water for 8 to 12 hours
Gloves are essential protective equipment and must be worn before a blood draw. If a patient has eaten recently (less than 2 hours
when venipunctures are performed. When gloves are removed, earlier), there will be a temporary increase in glucose and lipid
it is important that no substances from the soiled gloves come content in the blood. As a result, the serum or plasma may appear
in contact with the hands. Glove removal is covered in detail cloudy or turbid (lipemic), which interferes with testing, especially
in Chapter 2. for tests such as glucose level, sodium level, and complete blood
Contaminated sharps and infectious wastes should be placed counts.
in designated puncture-resistant containers. The red or red-
orange biohazard sign (see Figure 2-2) indicates that a container Smoking
holds potentially infectious materials. Biohazard containers Patients who smoke before blood collection may have increased
should be easily accessible and should not be overfilled. white blood cell counts and cortisol levels. Long-term smoking can
lead to decreased pulmonary function and result in increased hemo-
globin levels. Skin puncture samples may be more difficult to obtain
RESPONSIBILITY OF THE PHLEBOTOMIST as a result of impaired circulation.
IN INFECTION CONTROL
Because phlebotomists interact with patients and staff through-
out the day, they potentially can infect numerous people. Phle- (supine or erect), diurnal rhythms (day or night), exercise,
botomists should become familiar with and observe infection stress, diet (fasting or not), and smoking (Box 3-1).3-5 It is impor-
control and isolation policies. Violations of policies should tant that the phlebotomist adhere to requests for specimens to
be reported. A phlebotomist must maintain good personal be drawn at a specific time and record the time of collection.
health and hygiene, making sure to have clean clothes, clean
hair, and clean fingernails. Standard precautions must be fol-
VENIPUNCTURE
lowed at all times, with special attention to the use of gloves
and handwashing. This chapter offers only an overview of specimen collection;
texts that give detailed information are listed in the references.

PHYSIOLOGIC FACTORS AFFECTING


Collection Equipment for Venipuncture
TEST RESULTS
Tourniquet
Certain physiologic factors specific to the patient may affect A tourniquet is used to provide a barrier against venous blood
results of laboratory testing. These factors include posture flow to help locate a vein. A tourniquet can be a disposable
20 PART I Introduction to Hematology

elastic strap, a heavier Velcro strap, or a blood pressure cuff. evacuated tube holder or to be attached to the tips of syringes.
The tourniquet should be applied 2 to 4 inches above the Evacuated tube needles are double pointed and have a long,
venipuncture site and left on for no longer than 1 minute narrow, pointed end that punctures the patients skin and is
before the venipuncture is performed. Latex-free tourniquets covered by a plastic cap, and a short beveled point at the other
are available for individuals with a latex allergy. end that punctures the rubber stopper of the evacuated tube
and is covered by a rubber sleeve. The rubber sleeve prevents
Collection Tubes blood from dripping into the holder when tubes are changed
The most common means of collecting blood specimens is (Figure 3-2). On syringes, single-sample needles are used.
through the use of an evacuated tube system. The system The end of the needle that is inserted into the vein has a point
includes a tube, which can be either plastic or glass; a needle; with a slanted side (bevel), which must be facing up when the
and an adapter, which is used to secure the needle and the needle is inserted. Needle tips should be examined for burrs
tube. For safety, OSHA recommends the use of plastic tubes or bends before a venipuncture is performed. Gauge numbers
whenever possible. Most glass tubes are coated with silicone are related inversely to the bore size: the smaller the gauge
to help decrease the possibility of hemolysis and to prevent number, the larger the bore. Needle gauges for drawing blood
blood from adhering to the sides of the tube. All tubes come range from 20 gauge to 25 gauge. The most common needle
in various sizes and may contain a variety of premeasured size for adult venipuncture is 21 gauge with a length of 1 inch.
additives. The advantage of using a 1-inch needle is that it provides better
Although there are several manufacturers of evacuated tubes control.
in the United States all follow a universal color code in which
the stopper color indicates the type of additive contained Needle Holders
in the tube. Figure 3-1 provides a summary of collection Several new needles and holders have been designed to comply
tube types. with the revised Occupational Exposure to Bloodborne Patho-
gens Standard (effective April 18, 2001) and its requirement
Additives in Collection Tubes for implementation of safer medical devices. These needle
Antiglycolytic agent. An antiglycolytic agent inhibits the holders have safety features to prevent the possibility of needle
use of glucose by blood cells. Such inhibition may be necessary sticks. Needle holders are made to fit a specific manufacturers
if testing for glucose level is delayed. Examples of antiglycolytic needles and tubes and, for best results, should not be inter-
agents are sodium fluoride and lithium iodoacetate. Tubes changed. The holders are disposable and must be discarded
containing sodium fluoride alone yield serum. Sodium fluo- after a single use with the needle still attached as per OSHA
ride is combined with potassium oxalate or potassium ethyl- requirements.6
enediaminetetraacetic acid (K2EDTA), both anticoagulants, to Examples of these needles and holders are the following:
yield plasma for more rapid testing. 1. The Vacutainer Eclipse Blood Collection System (Becton,
Anticoagulant agent. An anticoagulant prevents blood Dickinson and Company, Franklin Lakes, N.J.) allows
from clotting. The mechanism by which clotting is prevented single-handed activation after the venipuncture is per-
varies with the anticoagulant. EDTA, citrate, and oxalate formed by pushing the safety shield forward with the thumb
remove calcium by forming insoluble salts, whereas heparin until an audible click is heard. The Becton Dickinson Eclipse
prevents the conversion of prothrombin to thrombin. If needle should be used with a single-use tube holder. After
calcium is removed or thrombin is not formed, coagulation the safety shield is activated, the entire assembly should be
does not occur. Examples of anticoagulants are EDTA, sodium discarded intact into the sharps container.
citrate, and lithium or sodium heparin. Tubes must be inverted 2. The Jelco multisample blood collection needle used with
gently several times to ensure proper mixing immediately after the Venipuncture Needle-Pro Device (Smiths Medical ASD,
collection, according to the manufacturers instructions. Norwell, Mass.) allows the Needle-Pro sheath to be snapped
Clot activator. A clot activator helps initiate or enhance over the needle by pushing it against a flat, firm surface after
the clotting mechanism. Clot activators include glass or silica the venipuncture is completed. The entire device is disposed
particles that provide increased surface area for platelet activa- of into the sharps container (Figure 3-3).
tion and a clotting factor such as thrombin. 3. Greiner Bio-One (Monroe, N.C.) offers the VACUETTE
Separator gel. Separator gel is an inert material that QUICKSHIELD, which has a sheath that locks into place
undergoes a temporary change in viscosity during the centrifu- after use, and the QUICKSHIELD Complete PLUS, a system
gation process, which enables it to serve as a separation barrier that incorporates a holder with the VACUETTE Visio PLUS
between the liquid (serum or plasma) and cells. Because this multisample needle attached. The flash window in the
gel may interfere with some testing, serum or plasma from needle hub indicates when a successful venipuncture has
these tubes cannot be used with certain instruments or for been achieved (Figure 3-4).
blood bank procedures.
Winged Infusion Sets (Butterflies)
Needles A butterfly is an intravenous device that consists of a short
Sterile needles come in a variety of lengths and gauges (bore needle and a thin tube with attached plastic wings (Figure 3-5).
or opening size). Needles are made to be screwed into the The butterfly can be connected to evacuated tube holders,
CHAPTER 3 Specimen Collection 21

BD Vacutainer Venous Blood Collection

Tube Guide
For the full array of BD Vacutainer Blood Collection Tubes, visit www.bd.com/vacutainer.
Many are available in a variety of sizes and draw volumes (for pediatric applications). Refer to our website for full descriptions.

BD Vacutainer Tubes BD Vacutainer Tubes Inversions
with with at Blood Your Labs

BD Hemogard Closure Conventional Stopper Additive Collection* Laboratory Use Draw Volume/Remarks
Clot activator and gel 5 For serum determinations in chemistry.
for serum separation May be used for routine blood donor
Red/ screening and diagnostic testing of serum
Gold
Gray for infectious disease.** Tube inversions
ensure mixing of clot activator with blood.
Blood clotting time: 30 minutes.
Lithium heparin 8 For plasma determinations in chemistry.
and gel for plasma Tube inversions ensure mixing of anticoagulant
Light Green/ separation (heparin) with blood to prevent clotting.
Green Gray

Silicone coated (glass) 0 For serum determinations in chemistry.


Clot activator, Silicone 5 May be used for routine blood donor
coated (plastic) screening and diagnostic testing of serum
Red Red
for infectious disease.** Tube inversions
ensure mixing of clot activator with blood.
Blood clotting time: 60 minutes.
Thrombin 8 For stat serum determinations in chemistry.
Tube inversions ensure mixing of clot activator
Gray/ (thrombin) with blood to activate clotting.
Orange
Yellow

Clot activator 8 For trace-element, toxicology, and


(plastic serum) nutritional-chemistry determinations.
K2EDTA (plastic) 8 Special stopper formulation provides
Royal
low levels of trace elements
Blue
(see package insert). Tube inversions ensure
mixing of either clot activator or anticoagulant
(EDTA) with blood.

Sodium heparin 8 For plasma determinations in chemistry.


Lithium heparin 8 Tube inversions ensure mixing of anticoagulant
Green Green (heparin) with blood to prevent clotting.

Potassium oxalate/ 8 For glucose determinations. Oxalate and


sodium fluoride EDTA anticoagulants will give plasma
Sodium fluoride/Na2 EDTA 8 samples. Sodium fluoride is the
Gray Gray
Sodium fluoride 8 antiglycolytic agent. Tube inversions
(serum tube) ensure proper mixing of additive with blood.

K2EDTA (plastic) 8 For lead determinations. This tube is certified


to contain less than .01 g/mL(ppm) lead.
Tan Tube inversions prevent clotting.

Sodium 8 SPS for blood culture specimen collections


polyanethol sulfonate (SPS) in microbiology.
Acid citrate dextrose
additives (ACD): ACD for use in blood bank studies, HLA
Solution A - 8 phenotyping, and DNA and paternity testing.
Yellow
22.0 g/L trisodium citrate,
8.0 g/L citric acid, 24.5 g/L Tube inversions ensure mixing of anticoagulant
dextrose with blood to prevent clotting.
Solution B - 8
13.2 g/L trisodium citrate,
4.8 g/L citric acid, 14.7 g/L
dextrose
Liquid K3EDTA (glass) 8 K2EDTA and K3EDTA for whole blood
Spray-coated K2EDTA 8 hematology determinations. K2EDTA may be
(plastic) used for routine immunohematology testing,
Lavender Lavender
and blood donor screening.***
Tube inversions ensure mixing of anticoagulant
(EDTA) with blood to prevent clotting.
K2EDTA with gel 8 For use in molecular diagnostic test methods
(such as, but not limited to, polymerase chain
reaction [PCR] and/or branched DNA [bDNA]
White
amplification techniques.) Tube inversions
ensure mixing of anticoagulant (EDTA) with
blood to prevent clotting.
Spray-coated K2EDTA 8 For whole blood hematology determinations.
(plastic) May be used for routine immunohematology
testing and blood donor screening.***
Pink Pink
Designed with special cross-match label for
patient information required by the AABB.
Tube inversions prevent clotting.
Buffered sodium citrate 3-4 For coagulation determinations. CTAD for
0.105 M (3.2%) glass selected platelet function assays and routine
Light Light 0.109 M (3.2%) plastic coagulation determination. Tube inversions
Blue Blue ensure mixing of anticoagulant (citrate) to
Citrate, theophylline, 3-4
adenosine, dipyridamole prevent clotting.
(CTAD)

Clear

None (plastic) 0 For use as a discard tube or secondary


New specimen tube.
Clear
Red/
Light Gray


Note: BD Vacutainer Tubes for pediatric and partial draw applications can be found on our website.
BD Diagnostics BD Global Technical Services: 1.800.631.0174 * Invert gently, do not shake
** The performance characteristics of these tubes have not been established for infectious disease testing in general; therefore, users must
Preanalytical Systems vacutainer_techservices@bd.com validate the use of these tubes for their specific assay-instrument/reagent system combinations and specimen storage conditions.
1 Becton Drive BD Customer Service: 1.888.237.2762 *** The performance characteristics of these tubes have not been established for immunohematology testing in general; therefore, users must
Franklin Lakes, NJ 07417 USA www.bd.com/vacutainer validate the use of these tubes for their specific assay-instrument/reagent system combinations and specimen storage conditions.

BD, BD Logo and all other trademarks are property of Becton, Dickinson and Company. 2008 BD Printed in USA 08/08 VS5229-9

Figure 3-1 Vacutainer tube guide. (Courtesy and Becton, Dickinson and Company.)
22 PART I Introduction to Hematology

Figure 3-2 Multisample needle. The rubber sleeve prevents blood from dripping into the holder when tubes are changed. (Courtesy and Becton,
Dickinson and Company.)

Figure 3-4 QUICKSHIELD Complete PLUS with flash window. Blood in


the flash window indicates successful venipuncture. (Courtesy Greiner
Bio-One GmbH.)
1 2 3
B
Figure 3-3 A, Jelco Needle-Pro. B, Use of Jelco Needle-Pro. (1) Attach
needle. (2) Remove cap and draw blood from patient. (3) Press sheath on
flat surface. (Courtesy Smiths Medical ASD, Norwell Mass.)

syringes, or blood culture bottles with the use of special adapt-


ers. Butterflies are useful in collecting specimens from children
or other patients from whom it is difficult to draw blood.
Butterflies now come with resheathing devices to minimize the
risk of needle stick injury (e.g., MONOJECT ANGEL WING
Blood Collection [Covidien, Mansfield, Mass.], Vacutainer
brand SAFETY-LOK and Vacutainer Push Button Blood Collec-
tion Set [Becton Dickinson], and VACUETTE Safety Blood
Collection Set [Greiner Bio-One]).

Syringes
Syringes consist of a barrel that is graduated in milliliters and Figure 3-5 Butterfly Saf-T Wing. (Courtesy Smiths Medical ASD,
a plunger. Syringe needles have a point at one end and an open Norwell, Mass.)
CHAPTER 3 Specimen Collection 23

center and working outward. It is important to allow the area


Saf-T Holder
to air-dry before the venipuncture is performed so that the
patient does not experience a burning sensation and contami-
nation of the specimen is prevented. When a sterile site is
prepared for collection of specimens for blood culture, a two-
step procedure is used in which cleansing with isopropyl
alcohol is followed by cleansing with iodine. Some healthcare
Sample
Side clamp syringe facilities use a one-step application of chlorhexidine gluconate/
directs blood flow Luer isopropyl alcohol. To avoid contamination when legal blood
alcohol level is to be measured, benzalkonium chloride
(Zephiran) or a nonalcohol antiseptic is used.

A Selection of a Vein for Routine Venipuncture


The superficial veins of the antecubital fossa (bend in the
elbow) are the most common sites for venipuncture. The three
primary veins that are used are (1) the cephalic vein, located
on the upper forearm on the thumb side of the hand; (2) the
basilic vein, located on the inside (medial) aspect of the ante-
cubital fossa; and (3) the median cubital vein, which connects
the basilic and cephalic veins in the antecubital fossa. The
median cubital vein is the vein of choice (Figure 3-7).
If necessary, the phlebotomist should have the patient make

a fist after application of the tourniquet; the vein should
become prominent. The patient should not do any pumping
of the fist, because it may affect some of the test values. The
B
phlebotomist should palpate (examine by touching) the vein
Figure 3-6 A, Jelco closed blood collection system. (Courtesy Smiths with his or her index finger to determine vein depth, direction,
Medical ASD, Norwell, Mass.) B, Device for transferring blood from syringe and diameter. If a vein cannot be located in either arm, it may
to vacuum tube. (1) Draw sample with syringe. (2) Close clamp. (3) Insert
be necessary to examine the veins on the dorsal side of the
tube to transfer blood from sample syringe to tube. To fill additional tubes,
wrist and hand.
open clamp, draw syringe again, close clamp, and transfer. (Courtesy Smiths
Medical ASD, Norwell, Mass.) The veins in the feet should not be used without physician
permission. The policy in some institutions is to request that
a second phlebotomist attempt to locate a vein in the arm
before a vein in one of these three alternate sites is used.

hub at the other end that attaches to the barrel. Syringes come Routine Venipuncture Procedure
with different types of needle attachments and in different The phlebotomist should practice standard precautions, which
sizes. It is important to attach the needle securely to the syringe include applying gloves and washing hands at the beginning of
to prevent air from entering the system. Syringes may be useful the procedure and removing gloves and washing hands at the
in drawing blood from pediatric, geriatric, or other patients end of the procedure. The following steps are recommended by
with tiny, fragile, or rolling veins that would not be able to the Clinical and Laboratory Standards Institute (CLSI)4:
withstand the vacuum pressure from evacuated tubes. With a 1. Prepare the accession order.
syringe, the amount of pressure exerted is controlled by the 2. Identify the patient by having the patient verbally state his
phlebotomist. Syringes may also be used with winged infusion or her name and confirm with one of the patients unique
sets. If only one tube of blood is needed, the syringe needle is identification numbers (i.e., medical records number,
shielded, removed, and discarded in a sharps container, and a birth date, or Social Security number).
syringe blood transfer device (Becton Dickinson) or Saf-T 3. Verify that any dietary restrictions have been met (e.g.,
Holder with male Luer adapter (Smiths Medical ASD) is fasting, if appropriate) and check for any sensitivity to
attached to the syringe and a vacuum tube is inserted into the latex.
transfer device. The blood is transferred from the syringe into 4. Assemble supplies.
the tube using the tubes vacuum (Figure 3-6). 5. Reassure the patient.
6. Position the patient.
Solutions for Skin Preparation 7. Verify paperwork and tube selection.
The most common skin cleanser is 70% isopropyl alcohol. It 8. If necessary, to help in locating the vein, ensure that the
can be applied using a commercially prepared alcohol pad or patients hand is closed.
a cotton ball or piece of gauze soaked in alcohol. The site 9. Select an appropriate venipuncture site, giving priority to
should be cleaned with a circular motion, beginning in the the median cubital vein, and put on gloves.
24 PART I Introduction to Hematology

Cephalic
vein
Median Antecubital fossa
cubital
Anterior

Basilic
vein
Cephalic
Posterior
vein

Basilic
vein

Figure 3-7 Veins of the forearm (two views).

10. Cleanse the venipuncture site with 70% isopropyl alcohol, 19. Bandage the venipuncture site after checking to ensure that
working in concentric circles from the inside to outside. bleeding has stopped.
Allow skin to air-dry. 20. If a syringe has been used, fill the tubes using a syringe
11. Apply the tourniquet 2 to 4 inches above the selected transfer device.
puncture site for no longer that 1 minute. 21. Dispose of the puncture equipment and other biohazard-
12. Inspect the needle and equipment. ous waste.
13. Perform the venipuncture by anchoring the vein with the 22. Label the tubes with the correct information. The minimal
thumb 1 to 2 inches below the site and inserting the needle, amount of information that must be on each tube is as
bevel up, with a 15- to 30-degree angle between the needle follows:
and the skin. Collect tubes using the correct order of draw, a. Patients full name
and invert each tube containing any additive immediately b. Patients unique identification number
after collection. A particular order of draw is recommended c. Date of collection
when drawing multiple specimens from a single venipunc- d. Time of collection (military time)
ture. Its purpose is to avoid possible test result error e. Collectors initials or code number
because of cross-contamination from tube additives. The NOTE: Compare the labeled tube with the patients identi-
recommended order of draw is as follows: fication bracelet or have the patient verify that the infor-
a. Blood culture or sterile tubes (i.e., yellow stopper) mation on the labeled tube is correct whenever possible.
b. Coagulation tube (i.e., light blue stopper) 23. Carry out any special handling requirements (i.e., chilling
c. Serum tube with or without clot activator or gel (i.e., or protecting from light).
red, gold, or red-gray marbled stopper) 24. Cancel any phlebotomy-related dietary restrictions and
d. Heparin tubes (i.e., green or light green stopper) thank the patient.
e. EDTA tubes (i.e., lavender stopper) 25. Send the properly labeled specimens to the laboratory.
f. Oxalate/fluoride tubes (i.e., gray stopper) The most crucial step in the process is patient identification.
14. Release and remove the tourniquet as soon as blood flow The patient must verbally state his or her name, or someone
is established or after no longer than 1 minute. must identify the patient for the phlebotomist. A hospitalized
15. Ensure that the patients hand is open. patient also must be identified by his or her identification
16. Place gauze lightly over the puncture site without pressing bracelet. The patients name and unique identification number
down. must match the information on the test requisition. Any dis-
17. After the last tube has been released from the back of the crepancies must be resolved before the procedure can continue.
multisample needle, remove the needle and activate the Failure to confirm proper identification can result in a life-
safety device as per manufacturers directions. threatening situation for the patient and possible legal ramifica-
18. Apply direct pressure to the puncture site. tions for the facility. All tubes should be labeled immediately
CHAPTER 3 Specimen Collection 25
after the blood specimen has been drawn, with the label attached If the patient begins to faint, the phlebotomist should
to the tube before the phlebotomist leaves the patients side. remove the needle immediately, lower the patients head, and
apply cold compresses to the back of the patients neck and
Coagulation Testing loosen any constrictive clothing. The patient should take some
If only a coagulation tube is to be drawn for determination of deep breaths and be offered some orange juice or cold water
prothrombin time or activated partial thromboplastin time, to drink. The patient should sit for at least 30 minutes before
the first tube drawn may be used for testing. It is no longer leaving. The incident always should be documented.
necessary to draw a 3-mL discard into a nonadditive tube
before collecting for routine coagulation testing. Tubes for Hematoma
coagulation testing must be filled to the correct level to main- A hematoma results when leakage of a large amount of fluid
tain a 9:1 ratio of blood to anticoagulant to ensure accurate around the puncture site causes the area to swell. If swelling
test results. Underfilling of tubes results in prolonged test begins, the needle should be removed immediately and pres-
values. When a butterfly is used to draw a single blue-topped sure applied to the site for at least 2 minutes. Hematomas may
tube, a nonadditive tube or another light blue tube must be result in bruising of the patients skin around the puncture site
used to clear the dead space in the tubing before collecting the if adequate pressure is not maintained. Blood that leaks out of
tube to be used for testing. For special coagulation testing, the vein under the patients skin may clot and result in nerve
however, the second or third tube drawn should be used.4 compression and permanent damage to the patients arm. A
hematoma most commonly occurs when the needle goes
Venipuncture Procedure in Children through the vein, when the bevel of the needle is only partially
and Infants in the vein, and when the phlebotomist fails to apply enough
Pediatric phlebotomy requires experience, special skills, and a pressure after venipuncture.
tender touch. Excellent interpersonal skills are needed to deal
with distraught parents and with crying, screaming, and scared Failure to Draw Blood
children. Ideally, only experienced phlebotomists should draw One reason for failure to draw blood is that the vein is missed,
blood from children; however, the only way to gain experience often because of improper needle positioning. The needle
is through practice. Through experience, one learns what works should be inserted completely into the vein with the slanted
in different situations. Frequently, smaller-gauge (23-gauge or side (bevel) up, at an angle of 15 to 30 degrees. Figure 3-8
25-gauge) needles are employed. Use of syringes or butterflies shows reasons for unsatisfactory flow of blood. It is sometimes
may be advantageous with some infants veins. No matter what possible to enter the vein by redirecting the needle, but only
equipment is used, the real secret to a successful venipuncture an experienced phlebotomist should attempt this, because
is a compassionate assistant. The childs arm should be immo- such manipulation can cause discomfort and a disabling nerve
bilized as much as possible so that the needle can be inserted injury to the patient.
successfully into the vein and can be kept there if the child tries Occasionally, an evacuated tube has insufficient vacuum,
to move. Use of special stickers or character bandages as and insertion of another tube yields blood. Keeping extra tubes
rewards may serve as an incentive for cooperation; however, within reach during blood collection can avoid a recollection
the protocol of the institution with regard to their distribution when the problem is a technical issue associated with the tubes
must be followed. (i.e., inadequate vacuum).

Complications Encountered in Petechiae


Blood Collection Petechiae are small red spots indicating that small amounts of
Ecchymosis (Bruise) blood have escaped into the skin epithelium. Petechiae indi-
Bruising is the most common complication encountered in cate a possible coagulation problem and should alert the phle-
obtaining a blood specimen. It is caused by leakage of a small botomist to be aware of possible prolonged bleeding.
amount of fluid around the tissue. The phlebotomist can
prevent bruising by applying direct pressure to the venipunc- Edema
ture site, instead of having the patient bend the arm at the Swelling caused by an abnormal accumulation of fluid in the
elbow. intercellular spaces of the tissues is termed edema. The most
common cause is infiltration of the tissues by the solution
Syncope (Fainting) running through an incorrectly positioned intravenous cathe-
Fainting is the second most common complication encoun- ter. Edematous sites should be avoided for venipuncture
tered. Before drawing blood, the collector always should ask because the veins are hard to find and the specimens may
the patient whether he or she has had any prior episodes of become contaminated with the tissue fluid.
fainting during or after blood collection. The CLSI recom-
mends that ammonia inhalants no longer be used because they Obesity
may trigger an asthma attack or a sudden, exaggerated response In obese patients, veins may be neither readily visible nor easy
that could lead to patient injury. The phlebotomist should to palpate. Sometimes the use of a blood pressure cuff can aid
follow the protocol at his or her facility. in locating a vein. The cuff should not be inflated any higher
26 PART I Introduction to Hematology

Skin Vein Skin Vein Hematoma Skin Hematoma Vein

A Correct needle position B Needle inserted through vein C Partial needle insertion

Vein Skin Skin Veins Skin Vein

D Bevel resting on vein wall E Needle too near vein valve F Collapsed vein
Figure 3-8 Proper and improper needle insertion for venipuncture.

than the patients diastolic pressure and should not be left on tube too hard; or if contamination by alcohol or water occurs
the arm for longer than 1 minute. It is not advisable to probe at the venipuncture site or in the tubes. Hemolysis also can
blindly in the patients arm because muscle or nerve damage occur physiologically as a result of hemolytic anemias or severe
may result. renal problems. Testing hemolyzed specimens can alter test
results, such as levels of potassium and enzymes, which can
Intravenous Therapy have a negative impact on patient outcome.
Drawing blood from an arm with an intravenous catheter
should be avoided if possible. The arm opposite the arm with Burned, Damaged, Scarred, and Occluded Veins
the intravenous line should be used. If there is no alternative, Burned, damaged, scarred, and occluded veins should be
blood should be drawn below the catheter with the tourniquet avoided because they do not allow the blood to flow freely and
placed below the catheter site. It is preferable to have the nurse may make it difficult to obtain an acceptable specimen.
stop the infusion for 2 minutes before the specimen is drawn.
The CLSI recommends that 5mL of blood be drawn for discard Seizures and Tremors
before samples to be used for testing are obtained. It is impor- Patients occasionally experience seizures because of a preexist-
tant to note on the requisition and the tube that the specimen ing condition or as a response to the needle stick. If a seizure
was obtained from an arm into which an intravenous solution occurs, the needle should be removed immediately. The
was running.2,4 The phlebotomist always should follow the patients safety should be ensured by preventing injury from
protocol established at his or her facility. nearby objects.

Hemoconcentration Vomiting and Choking


Hemoconcentration is an increased concentration of larger If the patient begins vomiting, the patients head must be posi-
molecules and analytes in the blood as a result of a shift in tioned so that he or she does not aspirate any vomit. During
water balance. Hemoconcentration can be caused by leaving the vomiting or choking, the phlebotomist should prevent the
tourniquet on the patients arm for too long. It is recommended patient from hitting his or her head.
that the tourniquet not remain on for longer than 1 minute
before venipuncture. If it is left on for a longer time because of Allergies
difficulty in finding a vein, it should be removed for 2 minutes Some patients may be allergic to skin antiseptic substances
and reapplied before the venipuncture is performed.5 other than alcohol. Adhesive bandages and tape also may cause
an allergic reaction. Hypoallergenic tape should be used or
Hemolysis pressure applied manually until the bleeding has stopped com-
The rupture of red blood cells (RBCs) with the consequent pletely. Sensitivity to latex should be determined before any
escape of hemoglobina process termed hemolysiscan cause phlebotomy procedure.
the plasma or serum to appear pink or red. Hemolysis can
occur if too small a needle was used during a difficult draw; if Mastectomy Patients
the phlebotomist pulls back too quickly on the plunger of a The CLSI requires physician permission before blood is drawn
syringe, forces blood into a tube from a syringe, or shakes a from the same side as a prior mastectomy (removal of the
CHAPTER 3 Specimen Collection 27

breast), even in the case of bilateral mastectomies. The pressure Puncture across
on the arm that is on the same side as the mastectomy from a fingerprints
tourniquet or blood pressure cuff can lead to pain or lym-
phostasis from accumulating lymph fluid. The other arm
should be used whenever possible.

Inability to Obtain a Blood Specimen


Each institution should have a policy covering proper proce-
dure when a blood specimen cannot be collected. If two unsuc-
cessful attempts at collection have been made, the CLSI
recommends that the phlebotomist seek the assistance of
another caregiver with blood collection expertise. Another
Medial
individual can make two attempts to obtain a specimen. If side
a second person is unsuccessful, the physician should be Lateral of heel
notified. side of
heel
The patient has the right to refuse to give a blood specimen. Posterior
If gentle urging does not persuade the patient to allow blood tibial artery
to be drawn, the phlebotomist should alert the nurse, who A B
will either talk to the patient or notify the physician. The Figure 3-9 Areas for skin puncture specimens: heel (A) and finger (B).
phlebotomist must not try to force an uncooperative patient
to have blood drawn; it can be unsafe for the phlebotomist
and for the patient. In addition, forcing a patient of legal
age and sound mind to have blood drawn against his or her the lancets could cause serious injury to the bones of the
wishes can result in charges of assault and battery or unlawful fingers. The site of choice in infants is the lateral (outside) or
restraint. medial (inside) surface of the plantar side (bottom) of the heel,
If the patient is a child and the parents offer to help hold although there have been some problems with puncturing the
the child, it is usually all right to proceed. Any refusals or posterior tibial artery when the medial heel surface is used
problems should be documented for legal reasons. If the (Figure 3-9, A). In older children and adults, the palmar surface
patient is not in his or her room, the absence should be of the distal portion of the third (middle) or fourth (ring)
reported to the nursing unit so that the nurses are aware that finger on the nondominant hand may be used.7 The puncture
the specimen was not obtained. on the finger should be made perpendicular to the fingerprint
lines when a puncture device with a blade is used (Figure 3-9,
B). Warming can increase the blood flow sevenfold. The site
SKIN PUNCTURES
can be warmed with a warm washcloth or a commercial heel
Skin punctures often are performed in newborns; in pediatric warmer. The site should be warmed to a temperature no greater
patients younger than 1 year; in adults who are severely burned than 42 C for no longer than 2 to 5 minutes, unless the col-
and whose veins are being reserved for therapeutic purposes; lection is for capillary blood gas analysis. The skin puncture
and in elderly patients with fragile veins. When peripheral site should be cleansed with 70% isopropyl alcohol and
circulation is poor, however, accurate results may not be allowed to air-dry. Povidone-iodine should not be used because
obtained with specimens acquired by skin puncture. of possible blood contamination, which would produce falsely
Capillary blood is actually a mixture of venous blood, arte- elevated levels of potassium, phosphorus, or uric acid.
rial blood, and tissue fluid. When the puncture site is warmed,
the specimen more closely resembles arterial blood. Because Skin Puncture Technique
capillary specimens may generate slightly different test results, The finger or heel must be securely immobilized. Punctures
a notation should be made when the specimen is obtained by should not be made more than 2mm deep because of the risk
skin puncture.7 White blood cell counts in specimens obtained of bone injury and possible infection (osteomyelitis). For heel
by skin puncture may be 15% to 20% higher than the counts punctures on premature infants, it is advisable to use a punc-
in venous specimens.8 Clinically significantly higher glucose ture device with even less depth. Most devices on the market
values are found in specimens obtained by skin puncture com- for performing skin punctures come in varying depths. The use
pared with those obtained by venipuncture.7 This is especially of plastic tubes or Mylar-coated glass tubes is recommended
important to note when a glucose tolerance test is performed by OSHA to avoid broken glass and exposure to biohazardous
or when glucometer results are compared with findings from materials.
venous samples. The phlebotomist should not puncture an area that is
swollen or bruised or already has been punctured. The first
Collection Sites drop of blood should be wiped away to prevent contamination
In most patients, skin punctures may be performed on the heel of the specimen with tissue fluid and to facilitate the free flow
or finger. In infants, the finger should not be punctured because of blood.7
28 PART I Introduction to Hematology

10. Cleanse the puncture site with 70% isopropyl alcohol


using concentric circles, working from the inside to outside.
Allow skin to air-dry.
11. Perform the puncture. Puncture depth should not exceed
2mm.
12. Wipe away the first drop of blood. This removes any residual
alcohol and any tissue fluid contamination.
13. Make blood smears if requested.
14. Collect the specimens and mix as needed. If an insufficient
sample has been obtained because the blood flow has
stopped, repeat the puncture at a different site with all
new equipment. Order of collection of specimens is as
follows:
a. Tube for blood gas analysis
Figure 3-10 Examples of equipment used for collection of skin puncture b. Slides, unless made from sample in the EDTA micro
specimens. (Courtesy Dennis J. Ernst, MT[ASCP], Director, Center for collection tube
Phlebotomy Education, Inc.) c. EDTA microcollection tube
d. Other microcollection tubes with anticoagulants (i.e.,
Devices for Collecting Blood from green or gray)
Skin Puncture e. Serum microcollection tubes
Devices for collecting blood from skin puncture include capil- 15. Elevate the puncture site and apply pressure until bleeding
lary tubes and microcollection tubes.7 Capillary tubes of various has stopped.
sizes are available with or without heparin added. Microcollec- 16. Label the specimens with the required information.
tion tubes are available with or without additives, and the cap NOTE: Compare the labeled tube with the identification
colors on the tubes correspond with the colors on vacuum bracelet for inpatients; have outpatients verify that the
tubes. The order of drawing is different for microcollection information on the labeled tube is correct, whenever
tubes. The EDTA microcollection tube should be filled first to possible.
ensure adequate volume and accurate hematology results, 17. Handle the specimen appropriately.
especially for platelets, which tend to aggregate at the site of 18. Thank the patient and parents.
puncture. Other tubes containing anticoagulants should be 19. Dispose of all puncture equipment and biohazardous
collected next, followed by serum tubes.5 Labeling for capillary materials.
specimens should contain the same information as for vacuum 20. Complete paperwork and indicate skin puncture
tubes. Examples of skin puncture equipment are shown in collection.
Figure 3-10. 21. Deliver the properly labeled specimens to the laboratory.

Skin Puncture Procedure


PREPARATION OF BLOOD SMEARS
Standard precautions, which include applying gloves and
washing hands at the beginning of the procedure and removing Blood smears can be made directly from capillary blood
gloves and washing hands at the end of the procedure, should or from venous blood by the wedge or coverslip method. In
be implemented. The following steps are recommended by either method, the phlebotomist must remember to wipe
the CLSI7: away the first drop of blood and use the second drop to make
1. Obtain and examine the requisition form. the smear, if blood from a finger or heel stick is used (see
2. Assemble equipment and supplies. Chapter 15).
3. Greet the patient (and parents); identify the patient by
having the patient verbally state his or her name and
QUALITY ASSURANCE IN
confirm with patients identification number (i.e., medical
SPECIMEN COLLECTION
records number, birth date, or Social Security number).
4. Verify that any dietary restrictions have been met (e.g., To ensure accurate patient test results, it is essential that the
fasting), and check for any sensitivity to latex. blood collection process, which includes specimen handling,
5. Position the patient and the parents or designated holder be monitored. Patient diagnosis and medical care are based on
as necessary. the outcomes of these tests. The following areas should be
6. Put on gloves. monitored in specimen collection.
7. Organize equipment and supplies.
8. Select the puncture site. Technical Competence
9. Warm the puncture site. Warming increases the blood flow The individual performing phlebotomy should be trained
sevenfold. Use a commercial heel warmer or warm wash- properly in all phases of blood collection. Certification is
cloth (40 C to 42 C) for 2 to 5 minutes. recommended. Continuing education is encouraged to keep
CHAPTER 3 Specimen Collection 29
current on all the changes in the field. Competency should be
assessed and documented on an annual basis for each employee BOX 3-2 Reasons for Specimen Rejection
performing phlebotomy. The test order requisition and the tube identification do not match.
The tube is unlabeled, or the labeling, including patient identification
Collection Procedures number, is incorrect.
Periodic review of collection procedures is essential to main- The specimen is hemolyzed.
taining the quality of specimens. Proper patient preparation The specimen was collected at the wrong time.
and correct patient identification are crucial. The correct tube The specimen was collected in the wrong tube.
or specimen container must be used. The specimen was clotted, and the test requires whole blood.
The specimen was contaminated with intravenous fluid.
Anticoagulants and Preservatives The specimen is lipemic.*
The manufacturers instructions must be followed with regard *Lipemic specimens cannot be used for certain tests; however, the phlebotomist
to mixing of all tubes with additives to ensure accurate test has no control over this aspect. Collection of a specimen after patient fasting may
results and prevent formation of microclots in the tubes. All be requested to try to reduce the potential for lipemia.
tubes should be checked for cracks and expiration dates. The
additives should be observed for discoloration or cloudiness,
which could indicate contamination. New lot numbers of pressure cuff should be checked for leaks and accuracy. Centri-
tubes must be checked to verify draw and fill accuracy. When fuges should be maintained according to the manufacturers
blood is collected in the light blue tube for coagulation a 9:1 instructions for cleaning and timing verification.
ratio of blood to anticoagulant must be maintained to ensure
accurate results. Specimens must be stored and handled Reasons for Specimen Rejection
properly before testing. A laboratory result is only as good as the specimen provided.
At times it may be suspected that a specimen will not yield
Requirements for a Quality Specimen accurate results and must be rejected. Box 3-2 lists reasons for
Requirements for a quality specimen are as follows: specimen rejection.
1. Patient properly identified
2. Patient properly prepared for draw
SPECIMEN HANDLING
3. Specimens collected in the correct order and labeled
correctly Proper handling of specimens begins with the initiation of the
4. Correct anticoagulants and preservatives used test request and ends when the specimen is finally tested. Accu-
5. Specimens properly mixed by inversion, if required rate test results depend on what happens to the specimen
6. Specimens not hemolyzed during that time. This pretesting period is referred to as the
7. Specimens requiring patient fasting collected in a timely preanalytical phase of the total testing process.
manner Routine specimens must be adequately inverted to mix the
8. Timed specimens drawn at the correct time additive and blood. Shaking can result in hemolysis of the
specimen and lead to specimen rejection or inaccurate test
Blood Collection Attempts results. Specimens should be transported in an upright posi-
One individual should not attempt to obtain a specimen suc- tion to ensure complete clot formation and reduce agitation,
cessfully from a given patient more than twice. If two individu- which can result in hemolysis.
als have each tried twice without success, the physician should Exposure to light can cause falsely decreased values in tests
be contacted. There should be written procedures for what to measuring parameters such as bilirubin, carotene, RBC folate,
do when the patient is unavailable for a blood draw or when and urine porphyrin levels. For certain tests, the specimens
the patient refuses a draw. need to be chilled, not frozen, and should be placed in an ice-
water bath to slow down cellular metabolism. These tests
Collection of Specimens for Blood Culture include blood gas analysis, ammonia concentration, lactic acid
Each facility should monitor its blood culture contamination level, and certain coagulation tests. Other tests require that
rate and keep that rate lower than 3% as recommended by the specimens be kept warm to ensure accurate results. The cold
American Association of Microbiology.9-11 Failure to do so agglutinin test is one such test; if the specimen is refrigerated
could indicate a problem in the quality of all procedures being before the serum is removed, the antibody is reabsorbed onto
performed. the RBCs.
Most specimens for routine testing should be delivered to
Quality Control and Preventive Maintenance the laboratory within 45 minutes to 1 hour of collection for
for Specimen Collection Instruments processing. To ensure accurate results, less time is recom-
Thermometers used in refrigerators and freezers in which mended for tests such as those measuring glucose, potassium,
specimens are stored should be calibrated annually, or only cortisol, and some enzymes. The CLSI recommends that the
thermometers certified by the National Bureau of Standards maximum time limit for separating serum and plasma from
should be used. If bleeding times are to be measured, the blood cells be 2 hours from the time of collection.
30 PART I Introduction to Hematology

and skin antisepsis. No antiseptics containing alcohol should


LEGAL ISSUES IN PHLEBOTOMY
be used for skin antisepsis in such cases. Soap and water may
There are many daily practices in healthcare that, if performed be used if no other cleaners are available.
without reasonable care and skill, can result in a lawsuit. Facili- To minimize the risk of legal action, the phlebotomist
ties have been and will continue to be held legally accountable should do the following:
for the actions of those who collect blood for diagnostic testing. 1. Follow up on all incident reports.
Two areas of particular concern to phlebotomists are breach of 2. Participate in continuing education.
patient confidentiality and patient misidentification. Unless 3. Become certified in the profession.
there is a clinical need to know or a patient has given written 4. Acknowledge the extent of liability coverage.
permission, no one has a right to patient information. A patient 5. Follow established procedures.
will never be misidentified if correct procedures for specimen 6. Always exhibit professional, courteous behavior.
collection are followed. Phlebotomists often are called to 7. Always obtain proper consent.
testify in court in cases involving blood alcohol levels. The 8. Respect and honor the Patients Bill of Rights.
phlebotomist is asked about patient identification procedures 9. Maintain proper documentation.

SUMMARY
Laboratory test results are only as good as the specimen tested. CLSI guidelines should be followed for venipuncture and skin
Standard precautions must be followed in the collection of blood puncture.
to prevent exposure to bloodborne pathogens. Common complications of blood collection include bruising, faint-
Physiologic factors affecting test results include posture, diurnal ing, and hematoma.
rhythm, exercise, stress, diet, and smoking. Each institution should establish a policy covering proper proce-
Although there are several U.S. manufacturers of evacuated tubes, dure when a blood specimen cannot be obtained.
all follow a universal color coding system in which the stopper Following established procedures and documenting all incidents
color indicates the type of additive contained in the tube. minimize the risk of liability when performing phlebotomy.
The gauge numbers of needles relate inversely to bore size: the
smaller the gauge number, the larger the bore. Now that you have completed this chapter, go back and
The three primary veins used for phlebotomy are the cephalic, read again the case studies at the beginning and respond
basilic, and medial veins. to the questions presented.

R E V I E W Q UESTIONS
1. The vein of choice for performing a venipuncture is the: 5. What is the proper angle of needle insertion for
a. Basilic phlebotomy?
b. Cephalic a. 5 degrees
c. Median cubital b. 15 to 30 degrees
d. Femoral c. 35 degrees
d. 45 degrees
2. The most important step in phlebotomy is:
a. Cleansing the site 6. What is the recommended order of drawing when the
b. Identifying the patient evacuated tube system is used?
c. Selecting the proper needle length a. Gel separator, nonadditive, coagulation, and blood
d. Using the correct evacuated tube culture
b. Additive, nonadditive, gel separator, and blood
3. Failure to obtain blood by venipuncture may occur because culture
of all of the following except: c. Nonadditive, blood culture, coagulation, and other
a. Incorrect needle positioning additives
b. Tying the tourniquet too tightly d. Blood culture, coagulation, nonadditive, and gel
c. Inadequate vacuum in the tube separator or other additives
d. Collapsed vein
7. Acceptable sites for skin puncture on infants are:
4. The needle should be inserted into the arm with the bevel a. Middle of the heel
facing: b. Lateral or medial surface of the bottom of the heel
a. Down c. Inside of the heel, close to the arch of the foot, and
b. Up any of the toes, close to the tip
c. To either side d. Middle of the bottom of the heel
d. Makes no difference
CHAPTER 3 Specimen Collection 31

8. An anticoagulant is an additive placed in evacuated tubes What is your assessment of the phlebotomists technique?
to: a. All steps were performed acceptably in the proper
a. Make the blood clot faster order.
b. Dilute the blood before testing b. The phlebotomist should have labeled the tubes
c. Prevent the blood from clotting before collecting the specimen.
d. Ensure the sterility of the tube c. The phlebotomist should have cleansed the arm
while the tourniquet was in place.
9. You are evaluating a new phlebotomist on his venipunc- d. The phlebotomist should have removed the
ture performance. He completed the following steps in the tourniquet before removing the needle from the arm.
order listed:
Asked the outpatient his name and date of birth and 10. For a complete blood count (hematology) and measure-
compared those with the requisition ment of prothrombin time (coagulation), the phle
Applied a tourniquet and selected a prominent vein in botomist collected blood into a lavender-topped and a
the center of the antecubital fossa green-topped tube. Are these specimens acceptable?
Released the tourniquet, collected needed equipment, a. Yes, EDTA is used for hematologic testing and
and assembled it all heparin is used for coagulation testing.
Cleansed the venipuncture site and allowed it to dry b. No, although EDTA is used for hematologic testing,
Applied the tourniquet, unsheathed the needle, citrate, not heparin, is used for coagulation testing.
stretched the skin below the proposed venipuncture c. No, although heparin is used for hematologic testing,
site, and inserted the needle into the selected vein citrate, not EDTA, is used for coagulation testing.
within the cleansed area d. No, hematologic testing requires citrate and
Pushed the tube onto the holder while holding the coagulation testing requires a clot, so neither tube is
needle still, withdrew the tube from the holder, removed acceptable.
the needle from the arm, and engaged the safety device
Immediately applied a gauze pad to the site and released 11. Which step in the CLSI procedure for venipuncture is part
the tourniquet of standard precautions?
Applied pressure to the site while gently mixing the a. Wearing gloves
specimen b. Positively identifying the patient
Verified that the patient was not bleeding and applied c. Cleansing the site for the venipuncture
a bandage d. Bandaging the venipuncture site
Labeled the tube appropriately

REFERENCES
1. Rules and regulations: bloodborne pathogens. Fed Reg 7. Clinical and Laboratory Standards Institute: Procedures and
56:64175-64182, 1991. devices for the collection of diagnostic capillary blood specimens;
2. McCall RE, Tankersley CM: Phlebotomy essentials, ed 4, approved standard, ed 6, CLSI document H04-A6, Wayne, Pa,
Philadelphia, 2008, Lippincott Williams & Wilkins. 2008, Clinical and Laboratory Standards Institute.
3. Christensen RD, Hill HR: Exercise-induced changes in the 8. Geller J: Effect of sample collection on laboratory test results.
blood concentration of leukocyte populations in teenage ath- Paper presented at American Society of Clinical Pathologists
letes. Am J Pediatr Hematol Oncol 9:140-142, 1987. Spring 1992 Teleconference. Chicago, 1992.
4. Clinical and Laboratory Standards Institute: Procedures for the 9. Strand CL, Wajsbort RR, Sturmann K: Effect of iodophor vs.
collection of diagnostic blood specimens by venipuncture; approved tincture skin preparation on blood culture contamination
standard, ed 6, CLSI document H3-A6, Wayne, Pa, 2007, rate. JAMA 269:1004-1006, 1999.
Clinical and Laboratory Standards Institute. 10. Schifman RB, Strand CL, Meier FA, et al: Blood culture con-
5. Garza D, Becan-McBride K: Phlebotomy handbook, ed 7, Upper tamination: a College of American Pathologists Q-Probes
Saddle River, NJ, 2005, Pearson Prentice Hall. study involving 640 institutions and 497,134 specimens from
6. Occupational Safety and Health Administration (OSHA), US adult patients. Arch Pathol Lab Med 122:216-221, 1998.
Department of Labor: OSHA Safety and Health Information 11. Hall KK, Lyman JA: Updated review of blood culture con-
Bulletin (SHIB): Re-use of blood tube holders, October 15, 2003. tamination. Clin Microbiol Rev 19:788-802, 2006.

ADDITIONAL RESOURCES
Center for Phlebotomy Education, http://www.phlebotomy.com/. Lab Tests Online, http://www.labtestsonline.org/.
BD Vacutainers (Becton Dickinson). Available at: http://www.bd.
com/vacutainer/.
4 Care and Use of the Microscope
Bernadette F. Rodak

OUTLINE OBJECTIVES
Principles of Microscopy After completion of this chapter, the reader will be able to:
Component Parts and
Function of Each Part 1. Given either a diagram or an actual brightfield light focus a stained blood film with dry and oil immersion
Operating Procedure with microscope, identify the component parts. objectives.
Koehler Illumination 2. Explain the function of each component of a bright- 9. Describe the proper care and cleaning of micro-
Considerations field light microscope. scopes and recognize deviations from these
Immersion Oil and Types 3. Define achromatic, planachromatic, parfocal, and procedures.
Care of the Microscope parcentric as applied to lenses and microscopes; 10. Using the procedures described in the text and a
Basic Troubleshooting explain the advantages and disadvantages of microscope, properly clean the microscope after
Microlocator Slide each; and recognize examples of each from written routine use.
Other Microscopes Used descriptions of microscope use and effects. 11. Given the magnification of lenses in a compound
in the Clinical
4. Explain the purpose of adjusting microscope light microscope, calculate the total magnification.
Laboratory
using a procedure such as Koehler illumination. 12. Given a problem with focusing a blood film using a
Phase-Contrast
Microscope 5. List in proper order the steps for adjusting a bright- brightfield light microscope, suggest possible causes
Polarized Light field light microscope using Koehler illumination. and their correction.
Microscope 6. Using the procedure provided in the text and a 13. For each of the following, describe which compo-
Darkfield Microscope brightfield light microscope with appropriate compo- nents of the microscope differ from those of a
nents, properly adjust a brightfield light microscope standard light microscope, what the differences
by the use of Koehler illumination. accomplish, and what are the uses and benefits of
7. Describe the proper steps for viewing a stained blood each type in the clinical laboratory:
film with a brightfield light microscope, including use Phase-contrast microscope
of oil immersion lenses, and recognize deviations Polarized light microscope
from these procedures. Darkfield microscope
8. Using the procedure described in the text and a
brightfield light microscope with appropriate lenses,

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A Wright-stained peripheral blood film focuses under 10 objective. What steps should be taken to identify and correct
and 40 but does not come into focus under the 100 oil this problem?

M
icroscopes available today reflect improvement in ensure continued service from this powerful diagnostic instru-
every aspect from the first microscope of Anton van ment. The references listed at the end of this chapter address
Leeuwenhoek (1632-1723).1 Advanced technology the physical laws of light and illumination as applied to
as applied to microscopy has resulted in computer-designed microscopy.
lens systems, sturdier stands, perfected condensers, and built-in
illumination systems. Microscopes can be fitted with multiple
PRINCIPLES OF MICROSCOPY
viewing heads for teaching or conferences, or they can be
attached to a computer to allow an object to be projected onto In the compound microscope, an intermediate image of the
a monitor or a large screen. Regular care and proper cleaning illuminated specimen is formed by the objective lens in the

32
CHAPTER 4 Care and Use of the Microscope 33

standard microscopes, the brightfield illumination system,


Retina which passes light directly through the transparent specimen,
is used.

COMPONENT PARTS AND FUNCTION


OF EACH PART
Component parts and the function of each part of the micro-
Lens of eye
scope are summarized as follows (Figure 4-2):
1. The oculars, or eyepieces, usually are equipped with 10
lenses (degree of magnification is 10). The lenses magnify
the intermediate image formed by the objective lens in the
optical tube; they also limit the area of visibility. Micro-
scopes may have either one or two adjustable oculars. All
Eyepiece oculars should be used correctly for optimal focus (see
section on operating procedure). Oculars should not be
interchanged with the oculars of other models of micro-
Image formed
by objective scopes. The oculars in a pair are optically matched.
2. The interpupillary control is used to adjust the lateral separa-
tion of the oculars for each individual. When it is properly
adjusted, the user should be able to focus both eyes com-
fortably on the specimen and visualize one clear image.
3. The optical tube connects the oculars with the objective
250 mm
(10 inches)
lens. The intermediate image is formed in this component.
The standard length is 160mm, which, functionally, is the
Objective lens distance from the real image plane (oculars) to the objec-
tive lenses.
4. The neck, or arm, provides a structural site of attachment
Specimen
for the revolving nosepiece.
5. The stand is the main vertical support of the microscope.
The stage assembly, together with the condenser and base,
Condenser
is supported by the stand.
6. The revolving nosepiece holds the objectives and allows for
easy rotation from one objective lens to another. The
working distance between the objectives and the slide
varies with the make and model of the microscope.
7. There are usually three or four objective lenses (Figure 4-3),
each with a specific power of magnification. Engraved on
the barrel of each objective lens is the power of magnifica-
Virtual image tion and the numerical aperture (NA). The NA is related
to the angle of light collected by the objective; in essence,
it indicates the light-gathering ability of the objective lens.
Functionally, the larger the NA, the greater the resolution
or the ability to distinguish between fine details of two
Figure 4-1 Compound microscope. (From Abramowitz M: The micro- closely situated objects.
scope and beyond, vol 1, Lake Success, NY, 1985, Olympus Corp, p 2. Four standard powers of magnification and NA used in
Reprinted courtesy Eastman Kodak Company, Rochester, NY.) the hematology laboratory are 10/0.25 (low power),
40/0.65 or 45/0.66 (high power, dry), 50/0.90 (oil
immersion), and 100/1.25 (oil immersion). The smaller
optical tube. This image is then magnified and viewed through the magnification, the larger the viewing field; the larger
the oculars (Figure 4-1). the magnification, the smaller the viewing field. Total mag-
An example of a simple microscope is a magnifying lens that nification is calculated by multiplying the magnification
enlarges objects that are difficult to view with the unaided eye. of the ocular by the magnification of the objective lens; for
Movie theater projection units incorporate this system example, 10 (ocular) multiplied by 100 (oil immersion)
efficiently. is 1000 total magnification.
The compound microscope employs two separate lens systems, Microscopes employed in the clinical laboratory are
the product of which produces the final magnification. In used with achromatic or planachromatic objective lenses,
34 PART I Introduction to Hematology

1. Eyepieces
or oculars 1. Eyepieces or
oculars
2. Interpupillary
control 2. Interpupillary
3. Optical tube control
3. Optical
tube
4. Neck/arm
6. Revolving
nosepiece
7. Objective
lens
8. Stage
10. Condenser
11. Aperture diaphragm
control lever
12. Stage
controls
9. Focus
controls
13. Field
diaphragm
14. Light
source
5. Stand

Figure 4-2 Components of a microscope. (Courtesy Commercial Imaging & Design, Inc., Royal Oak, Mich.)

NA 1.25
Magnification 100
Tube length 160

Figure 4-3 Microscope objective lens. NA, Numerical aperture. (Courtesy Commercial Imaging & Design, Inc., Royal Oak, Mich.)
CHAPTER 4 Care and Use of the Microscope 35

whose function is to correct for chromatic and spheric the main condenser, it should be out for use with the 4
aberrations. Chromatic aberrations are caused by the spheric objective lens and in for lenses with magnification of 10
surface of the lens, which acts as a prism. As the various and higher. If it is below the condenser, it should be in for
wavelengths pass through the lens, each focuses at a dif- use with the 4 objective lens and out for lenses of mag-
ferent point, which gives rise to concentric rings of color nification of 10 and higher. The 4 objective is not used
near the periphery of the lens. Spheric aberrations result as routinely for examination of peripheral blood films.
light waves travel through the varying thicknesses of the The stage and condenser (Figure 4-4) consist of (1)
lens, blurring the image. The achromatic objective lens brings a control lever for a swing-out lens, (2) an aperture
light of two colors into focus, partially correcting for the diaphragm control lever, (3) a control for vertical adjust-
aberrations. When achromatic objective lenses are used, ment of the condenser, and (4) a condenser aperture
the center of the field is in focus, whereas the periphery is diaphragm.
not. A planachromatic lens, which is more expensive, also 11. The control lever swings the condenser top lens out of
corrects for curvature of the field, which results in a flat position.
field with uniform focus.2 Planachromatic lenses some- 12. The stage controls located under the stage move it along an
times are referred to as flat field lenses. For critical micros- x- or a y-axis.
copy, a planapochromatic lens, which brings light of three 13. The field diaphragm is located below the condenser within
colors into focus and almost completely corrects for chro- the base. When it is open, it allows a maximally sized circle
matic aberration, may be used. This type of objective lens of light to illuminate the slide. Almost closing the dia-
is fairly expensive and is rarely needed for routine labora- phragm, when low power is used, assists in centering the
tory use. condenser apparatus by the use of two centering screws.
A set of lenses with corresponding focal points all in Some microscopes have permanently centered condensers,
the same plane is said to be parfocal. As the nosepiece is whereas in others the screws are used for this function. The
rotated from one magnification to another, the specimen glass on top of the field diaphragm protects the diaphragm
remains in focus, and only minimal fine adjustment is from dust and mechanical damage.
necessary. 14. Microscopes depend on electricity as the primary source
8. The stage supports the prepared microscope slide to be for illumination power. There are two types of brightfield
reviewed. A spring assembly secures the slide to the stage. illumination: (1) critical illumination, in which the light
9. The focus controls (or adjustments) can be incorporated source is focused at the specimen, which results in increased
into one knob or can be two separate controls. When a but uneven brightness; and (2) the Koehler (or Khler)
single knob is used, moving it in one direction engages system, in which the light source is focused at the con-
the coarse control, whereas moving it in the opposite denser aperture diaphragm. The end result of Koehler illu-
direction engages the fine control. One gradation interval mination is a field of evenly distributed brightness across
of turning is equivalent to 2m. Many microscopes the specimen. Tungsten-halogen light bulbs are used most
are equipped with two separate adjustments: one coarse frequently as the illumination source. They consist of a
and one fine. The order of usage is the same: engage the tungsten filament enclosed in a small quartz bulb that is
coarse adjustment first and then fine-tune with the fine filled with a halogen gas. Tungsten possesses a high
adjustment. melting point and gives off bright yellowish light. A blue
10. The condenser, consisting of several lenses in a unit, may (daylight) filter should be used to eliminate the yellow
be permanently mounted or vertically adjustable with a color produced by tungsten.4,5 The light control knob turns
rack-and-pinion mechanism. It gathers, organizes, and on the light and should be used to regulate the brightness
directs the light through the specimen. Attached to and at of the light needed to visualize the specimen. The aperture
the bottom of the condenser is the aperture diaphragm, an diaphragm should never be used for this purpose, because
adjustable iris containing numerous leaves that control the closing it reduces resolving ability.3
angle and amount of the light sent through the specimen.
The angle, also expressed as an NA, regulates the balance
OPERATING PROCEDURE WITH KOEHLER
between contrast (ability to enhance parts within a cell)
ILLUMINATION
and resolution (ability to differentiate fine details of two
closely situated objects). The best resolution is achieved The procedure outlined here applies to microscopes with a
when the iris is used fully open, but there is some sacrifice nonfixed condenser. The following steps should be performed
of image contrast. In practice, this iris is closed only at the start of each laboratory session using the microscope:
enough to create a slight increase in image contrast. 1. Connect the microscope to the power supply.
Closing it beyond this point leads to a loss of resolution. 2. Turn on the light source.
Some microscopes are equipped with a swing-out lens 3. Open all diaphragms.
immediately above or below the main condenser lens. 4. Revolve the nosepiece until the 10 objective lens is
This lens is used to permit a wider field of illumination directly above the stage.
when the NA of the objective lens is less than 0.25 (e.g., 5. Adjust the interpupillary control so that looking through
the 4/0.12 objective lens).3 If the swing-out lens is above both oculars yields one clear image.
36 PART I Introduction to Hematology

2. Aperture diaphragm
control lever
4. Condenser
diaphragm

1. Swing-out lens

3. Vertical adjustment
of condenser

Figure 4-4 Condenser. (Courtesy Commercial Imaging & Design, Inc., Royal Oak, Mich.)

6. Place a stained blood film on the stage and focus on it, completely. Reopen the condenser diaphragm until the
using the fixed ocular, while covering the other eye. (Do leaves just disappear from view. Replace the ocular.
not simply close the other eye, because this would neces- 14. Rotate the nosepiece until the 40 objective lens is above
sitate adjustment of the pupil when you focus with the the slide. Adjust the focus (the correction should be
other ocular.) minimal) and find the cell that you had centered. If it is
7. Using the adjustable ocular and covering the opposite eye, slightly off center, center it again with the stage x-y control.
focus on the specimen. Start with the ocular all the way Note the greater amount of detail that you can see.
out, and adjust inward. If using two adjustable oculars, 15. Move the 40 objective out of place. Place a drop of
focus each individually. immersion oil on top of the slide. Rotate the nosepiece
8. Raise the condenser to its upper limit. until the 100 objective lens is directly above the slide.
9. Focus the field so that the cells become sharp and clear. Avoid moving a nonoil immersion objective through the
Concentrate on one cell and place it in the center of drop of oil. Adjust the focus (the correction should be
the field. minimal) and observe the detail of the cell: the nucleus
10. Close the field (lower) diaphragm. Look through the and its chromatin pattern; the cytoplasm and its color and
oculars. A small circle of light should be seen. If the light texture. The objective lens should dip into the oil slightly.
is not in the center of the field, center it by using the two
centering screws located on the condenser. This step is Considerations
essential, because an off-center condenser will result in 1. When revolving the nosepiece from one power to another,
uneven distribution of light. Adjust the vertical height of rotate it in such a direction that the 10 and 40 objective
the condenser so that you see a sharp image of the field lenses never come into contact with the oil on a slide. If oil
diaphragm, ringed by a magenta halo. If the substage con- inadvertently gets onto the high dry objective, clean the
denser is raised too much, the halo is orange; if it is objective immediately.
lowered too far, the halo is blue. 2. Parcentric refers to the ability to center a cell in question in
11. Reopen the field diaphragm until the image is nearly at the microscopic field and rotate from one magnification
the edge of the field, and fine-tune the centering process. power to another while retaining the cell close to the center
12. Open the diaphragm slightly until the image just of the viewing field. Recentering of the cell at each step is
disappears. minimal. Most laboratory microscopes have this feature.
13. Remove one ocular and, while looking through the micro- 3. In general, when the 10 and 40 objective lenses are used,
scope (without the ocular), close the condenser diaphragm the light intensity should be low. When the 50 and 100
CHAPTER 4 Care and Use of the Microscope 37

objective lenses are used, increase the intensity of the light 5. When fresh oil is added to residual oil on the 100 objective
by adjusting only the light control knob or by varying neutral lens, there may be loss of contrast. Clean off all residual
density filters. Neutral density filters are used to reduce the oil first.
amplitude of light and are available in a variety of 6. Do not use water to clean lenses. Your condensed breath on
densities.3 the lens surface may be useful in cleaning slightly soiled
4. Do not change the position of the condenser or the aperture lenses.
lever to regulate light intensity. The condenser should 7. When transporting the microscope, place one hand
always be in its upward position. The aperture lever is used under the base as support and one hand firmly around
only to achieve proper contrast of the features of the speci- the arm.
men being viewed. In addition to daily care of the microscope, semiannual or
annual maintenance with thorough cleaning should be done
by a professional. Microscope professionals may recognize and
IMMERSION OIL AND TYPES
correct problems with mechanics or optics before they are
Immersion oil is required to increase the refractive index when detected by the microscope user. They can correct problems
either the 50 or the 100 oil immersion objective lens is used. such as sticking of stage controls or incorrect optical alignment
The refractive index is the speed at which light travels in air that can lead to physical problems like carpal tunnel syndrome
divided by the speed at which light travels through a substance. and headaches.
This oil, which has the same properties as glass, allows the
objective lens to collect light from a wide NA, which provides
BASIC TROUBLESHOOTING
high resolution of detail.
Three types of immersion oil, differing in viscosity, are Most common problems are related to inability to focus. Once
employed in the clinical laboratory: the operator has ensured that he or she is not trying to obtain
1. Type A has very low viscosity and is used in fluorescence and a flat field using an objective lens that is not planachromatic,
darkfield studies. the following checklist can aid in identifying the problem:
2. Type B has high viscosity and is used in brightfield and Oculars
standard clinical microscopy. In hematology, this oil is rou- Clean?
tinely used. Securely assembled?
3. Type C has very high viscosity and is used with inclined Objective lens
microscopes with long-focus objective lenses and wide con- Screwed in tightly?
denser gaps. Dry objective free of oil?
Bubbles in the oil tend to act as prisms and consequently Condenser
reduce resolution. Bubbles may be created when oil is applied Adjusted to proper height?
to the slide. They are caused by lowering the objective imme- Free of oil?
diately into the oil. Sweeping the objective from right to left in Slide
the oil eliminates bubbles.5 Correct side up?
Coverslip
Correct side of smear?
CARE OF THE MICROSCOPE
Only one coverslip on slide?
Care of the microscope involves the following details: Free of mounting media?
1. When not in use for an extended period of time, the micro- Light source
scope always should be covered or protected from dust. Fingerprints on bulb?
2. Before use, inspect the component parts. If dust is found, Bulb in need of changing?
use an air syringe, a camel hair brush, or a soft lint-free Light source aligned correctly?
cloth to remove it. Using lens paper directly on a dirty
lens without first removing the dust may scratch the lens.
MICROLOCATOR SLIDE
Do not use laboratory wipes or facial tissue to clean the
lenses.6 Some microscope stages include locator values for x and y on
3. Avoid placing fingers on the lens surface. Fingerprints affect the stage, which allows the operator to take coordinates of a
the contrast and resolution of the image. cell so that it can be located easily for review. When locator
4. Use solvent sparingly. The use of xylene is discouraged, values are not incorporated into the stage, a microlocator slide
because it contains a carcinogenic component (benzene). or microslide field finder can be used.
Xylene is also a poor cleaning agent, leaving an oily film on The field finder slide is a commercially manufactured stan-
the lens. Lens cleaner or 70% isopropyl alcohol employed dard glass slide with a precise etched coordinate grid running
sparingly on a cotton applicator stick can be used to clean along the x and y axes. Figure 4-5 shows one example of such
the objective lenses. Alcohol should be kept away from the a slide.
periphery of the lenses, because alcohol can dissolve the 1. When a cell of interest is located on a prepared slide, the
cement and seep into the back side of the lens. cell should be centered under a high dry or oil objective and
38 PART I Introduction to Hematology

This phase difference produces variation in light intensity


from bright to dark, creating contrast in the image. Often the
objects appear to have haloes surrounding them.
In hematology, phase-contrast microscopy is employed in
counting platelets in a hemacytometer, since they are difficult
to visualize and count using brightfield microscopy. It also
can be used to view formed elements in unstained urine
sediments.

Polarized Light Microscope


Polarized light microscopy is another contrast-enhancing tech-
nique used to identify substances such as crystals in urine and
other body fluids (see Chapter 17). With brightfield micro
Figure 4-5 Microlocator slide. scopy, light vibrates in all directions. If a polarizer (filter) is
placed in the light path, the light vibrates in only one direction
then under the 10 objective lens. Note whether the feathered or plane, which creates polarized light. To convert a brightfield
edge of the blood film faces right or left. microscope to a polarizing one, two filters are needed. One
2. Carefully remove the slide from the spring assembly, taking filter (the polarizer) is placed below the condenser and allows
care not to disturb the position of the stage. only light vibrating in the east-west direction perpendicular to
3. Place the microlocator slide onto the stage. the light path to pass through the specimen. The second filter
4. View the microlocator through the ocular and record the (the analyzer) is placed between the objective and the ocular
letter-number combination. and allows only light vibrating in a north-south direction to
5. To relocate the cell, place the microlocator onto the stage pass to the ocular. When the transmission axes of these two
and place the recorded letter-number combination in the filters are oriented at right angles, no light can pass through
center of the stage. the pair to the ocular. When polarized light (vibrating in an
6. Remove the locator slide carefully and replace it with the east-west direction) passes through an optically active sub-
initial blood film. The cell of interest should be in the field. stance such as a monosodium urate crystal, however, the light
is refracted into two beams, one vibrating in the original direc-
tion (east-west) and one vibrating in a plane 90 degrees to it
OTHER MICROSCOPES USED IN THE
(i.e., north-south). The refracted light vibrating in the north-
CLINICAL LABORATORY
south direction can pass through the second filter (the ana-
Phase-Contrast Microscope lyzer) and is visible at the ocular. The magnified crystal appears
The ability to view a stained specimen by the use of brightfield white against a black background. If a first-order red compensa-
microscopy is affected by two features: (1) the ability of the tor filter also is placed in the light path below the stage, the
specimen to absorb the light hitting it, and (2) the degree to background becomes pink-red, and the crystal appears yellow
which light waves traveling through the specimen remain in or blue depending on its physical orientation relative to the
incident light path (east-west). Some crystals can be specifically
phase ( ). identified based on their unique birefringent (doubly refrac-
tive) characteristics when polarizing microscopy is used (see
Specimens that are transparent or colorless, such as Figures 17-20 and 17-21).
unstained cells, are not clearly visualized with brightfield
microscopy. Phase-contrast microscopy, through the installa-
tion of an annular diaphragm in the condenser, together with a Darkfield Microscope
phase-shifting element, creates excellent contrast of a cell against Darkfield microscopy is a contrast-enhancing technique that
its surrounding background. employs a special condenser. The condenser sends light up
The principle of phase contrast is related to the index of toward the specimen in a hollow cone. Because of the high
refraction and the thickness of a specimen, which produce angle of this cone, none of the illuminating rays enter the
differences in the optical path. Light passing through a trans- objective lens. Without the specimen in place, the field would
parent specimen travels slightly slower than light that is un appear black because of the absence of light. When the speci-
obstructed. The difference is so small that it is not noticeable men is in place, and if fine detail exists in the specimen, light
to the viewer. When a transparent phase plate is placed into is diffracted in all directions. This diffracted light is picked up
the microscope, however, the change in phase can be increased by the objective lens and appears as bright detail on a black
to half a wavelength, which makes the otherwise transparent background. Darkfield microscopy is helpful in microbiology
in the identification of spirochetes. When combined with the
objective visible ( ). use of fluorochrome dyes, darkfield fluorescence microscopy
can be used to identify lymphocyte subsets.
CHAPTER 4 Care and Use of the Microscope 39

SUMMARY
The compound microscope, through the use of an objective lens light by retarding a fraction of the light waves, resulting in a dif-
in the optical tube, forms an intermediate image of the illuminated ference in phase. This allows transparent or colorless objects to
specimen. The image is then magnified and viewed through the become visible.
oculars. Polarizing microscopes use two polarizing lenses to cancel the
The NA, which is engraved on the objective lenses, designates the light passing through the sample. If the object is able to polarize
light-gathering ability of the lens. The higher the NA, the greater light, as are some crystals, the light passing through is rotated
the resolution. and becomes visible.
Achromatic lenses maintain the center of the field in focus, Darkfield microscopes use condensers that send light to the
whereas planachromatic lenses also correct for the curvature of sample a high angle, directing the light away from the objective
a field, providing uniform focus. lens. If the sample has fine detail, it causes the light to bend back
The condenser gathers the light and directs it across a specimen. toward the objective, which allows it to be viewed against an
Koehler illumination establishes a field of evenly distributed bright- otherwise dark background.
ness across the specimen.
Microscopes should be carefully handled and maintained. Solvents Now that you have completed this chapter, go back and
should not be used to clean lenses; lens cleaner or 70% isopropyl read again the case study at the beginning and respond
alcohol is recommended. to the question presented.
Phase-contrast microscopy relies on the effect of index of refrac-
tion and the thickness of the specimen; these two features affect

R E V I E W Q UESTIONS
1. Use of which of the following type of objective lens causes 5. The total magnification obtained when a 10 ocular and
the center of the microscope field to be in focus, whereas a 10 objective lens are used is:
the periphery is blurred? a. 1
a. Planachromatic b. 10
b. Achromatic c. 100
c. Planapochromatic d. 1000
d. Flat field
6. After a microscope has been adjusted for Koehler illu
2. Which of the following gathers, organizes, and directs light mination, light intensity should never be regulated by
through the specimen? using the:
a. Ocular a. Rheostat
b. Objective lens b. Neutral density filter
c. Condenser c. Koehler magnifier
d. Optical tube d. Condenser

3. After focusing a specimen by using the 40 objective, the 7. The recommended cleaner for removing oil from objec-
laboratory professional switches to a 10 objective. The tives is:
specimen remains in focus at 10. Microscopes with this a. 70% alcohol or lens cleaner
characteristic are described as: b. Xylene
a. Parfocal c. Water
b. Parcentric d. Benzene
c. Compensated
d. Parachromatic 8. Which of the following types of microscopy is valuable
in the identification of crystals that are able to rotate
4. Which objective has the greatest degree of color light?
correction? a. Compound brightfield
a. Achromatic b. Darkfield
b. Planapochromatic c. Polarizing
c. Bichromatic d. Phase-contrast
d. Planachromatic
40 PART I Introduction to Hematology

9. A laboratory science student has been reviewing a hema- 10. Darkfield microscopes create the dark field by:
tology slide using the 10 objective to find a suitable a. Using two filters that cancel each other out, one
portion of the slide for examination. He moves the 10 above and the other below the condenser
objective out of place, places a drop of oil on the slide, b. Angling the light at the sample so that it misses the
rotates the nosepiece so that the 40 objective passes objective unless something in the sample bends it
through the viewing position, and continues to rotate the backward
100 oil objective into viewing position. This practice c. Closing the condenser diaphragm entirely, limiting
should be corrected in which way? light to just a tiny ray in the center of the otherwise
a. The stage of a parfocal microscope should be lowered dark field
before the objectives are rotated. d. Using a light source above the sample and collecting
b. The 100 oil objective should be in place for viewing light reflected from the sample, rather than
before the oil is added. transmitted through the sample, so that when there is
c. The drop of oil should be in place and the 100 no sample in place, the field is dark
objective lowered into the oil, rather than swinging
the objective into the drop.
d. The objectives should be rotated in the opposite
direction so that the 40 objective does not risk
entering the oil.

REFERENCES
1. Asimov I: Understanding physics: light, magnetism, and electricity, 5. Centonze Frohlich V: Major components of the light micro-
London, 1966, George Allen & Unwin. scope, J Vis Exp 17, July 30, 2008. Available at: http://www.
2. Microscope terms. Available at: http://www.microsocpe.com/ jove.com/index/details.stp?ID=843. Accessed June 2, 2010.
microscope-terms-t-5.html. Accessed June 2, 2010. doi:10.3791/843.
3. Olympus Microscopy Resource Center: Anatomy of the 6. Centonze Frohlich V: Proper care and cleaning of the micro-
MicroscopeKhler Illumination. Available at: http://www. scope, J Vis Exp 18, August 11, 2008. Available at: http://www.
olympusmicro.com/primer/anatomy/kohler.html. Accessed jove.com/index/details.stp?ID=842. Accessed June 2, 2010.
June 2, 2010. doi:10.3791/842.
4. Microscope Illumination. Olympus Microscope primer.
Available at: http://micro.magnet.fsu.edu/primer/anatomy/
illumination.html. Accessed June 2, 2010.

ADDITIONAL RESOURCES
Nikon Microscopy U, http://www.microscopyu.com/ Gill GW: Khler illumination. Lab Med 36(9):530, 2005.
Brunzel NA: Microscopy. In Fundamentals of urine and body fluid
analysis, ed 2, Philadelphia, 2004, Saunders, pp 1-23.
Quality Assurance in Hematology and
Hemostasis Testing
5
George A. Fritsma

OUTLINE OBJECTIVES
Validation of a New or After completion of this chapter, the reader will be able to:
Modified Assay
Accuracy 1. Validate and document a new or modified laboratory 8. Perform internal quality control using controls and
Precision assay. moving averages.
Linearity 2. Define accuracy and calculate accuracy using linear 9. Participate in periodic external quality assessment.
Limits regression to compare to a reference. 10. Measure and publish assay clinical efficacy.
Analytical Specificity 3. Compute precision using standard deviation and 11. Compute receiver operating characteristic curves.
Levels of Laboratory coefficient of variation. 12. Periodically assess laboratory staff competence.
Assay Approval 4. Determine assay linearity using graphical represen- 13. Provide continuing education programs for labora-
Documentation and tations and transformations. tory staff.
Reliability 5. Discuss analytical limits and analytical specificity. 14. Prepare a quality assurance plan to control for pre-
Lot-to-Lot Comparisons
6. Recount Food and Drug Administration clearance analytical and postanalytical variables.
Development of the
levels for laboratory assays. 15. List the agencies that regulate hematology and
Reference Interval and
Therapeutic Range 7. Compute a reference interval and a therapeutic hemostasis quality.
Internal Quality Control range for a new or modified assay.
Controls
Moving Average ( X B ) of
CASE STUDY
the Red Blood Cell
Indices After studying the material in this chapter, the reader should be able to respond to the following
Delta Checks case study:
External Quality
Assessment On an 8:00 AM assay run, the results for three levels of a preserved hemoglobin control sample are
Measurement of Clinical 2g/dL higher than the upper limit of the target interval. The medical laboratory scientist reviews
Efficacy -check data on the last 10 patient results and notices that the results are consistently 1.8 to 2.2g/
Receiver Operating dL higher than results generated the previous day.
Characteristic Curve 1. What do you call the type of error detected in this case?
Assay Feasibility 2. Can you continue to analyze patient samples as long as you subtract 2g/dL from the
Laboratory Staff results?
Competence 3. What aspect of the assay should you first investigate in troubleshooting this problem?
Proficiency Systems
Continuing Education
Quality Assurance Plan:
Preanalytical and
Postanalytical
Agencies that Address
Hematology and
Hemostasis Quality

41
42 PART I Introduction to Hematology

For many analytes, laboratory scientists employ primary


BOX 5-1 Examples of Components of Quality standards to standardize assays and establish accuracy. A
Assurance primary standard is a material of known, fixed composition
Preanalytical variables: selection of assay relative to patient need; that is prepared in pure form, often by weighing on an analyti-
implementation of assay selection; patient identification and prepara- cal balance. The scientist dissolves the weighed standard in
tion; specimen collection technique; specimen collection apparatus an aqueous solution, prepares suitable dilutions, calculates
used; specimen transport, preparation, and storage; monitoring of the anticipated concentration in each dilution, and assigns the
specimen condition calculated concentrations to assay outcomes. For example, the
Analytical variables: laboratory staff competence; assay and instru- scientist may obtain pure glucose, weigh 100mg, dilute it in
ment selection; assay validation, including linearity, accuracy, preci- 100mL of buffer, and assay an aliquot of the solution using
sion, analytical limits, and specificity; internal quality control; external photometry. The resulting absorbance would then be assigned
quality assessment the value of 100mg/dL. The scientist may repeat this proce-
Postanalytical variables: accuracy in transcription and filing of dure using a series of four additional glucose solutions at 20,
results, content and format of laboratory report, narrative report, 60, 120, and 160mg/dL to produce a five-point standard
reference interval and therapeutic range, timeliness in communicat- curve. The curve may be re-assayed several times to generate
ing critical values, patient and physician satisfaction, turnaround means for each concentration. The assay is then employed on
time, cost analysis human plasma, with absorbance compared with the standard
curve to generate a result. The matrix of a primary standard
need not match the matrix of the patient specimen; the stan-

I
n clinical laboratory science, quality implies the ability to dard may be dissolved in an aqueous buffer, whereas the test
provide accurate, reproducible assay results that offer clini- specimen may be human serum or plasma.
cally useful information.1 Because physicians base 70% of To save time and resources, the scientist may employ a
their clinical decision making on laboratory results, the results secondary standard, perhaps purchased, that the vendor has
must be reliable. Reliability requires vigilance and effort on the previously calibrated to a primary standard. The secondary
part of all laboratory staff members, and this effort is often standard may be a preserved plasma preparation delivered at
directed by an experienced medical laboratory scientist who is a certified known concentration. The scientist merely thaws or
a quality assurance and quality control specialist. reconstitutes the secondary standard and incorporates it into
Of the terms quality control and quality assurance, quality the test series during validation or revalidation. Manufacturers
assurance is the broader concept, encompassing preanalytical, often match secondary standards as closely as possible to the
analytical, and postanalytical variables (Box 5-1).2 Quality control test samples matrix, for instance, plasma to plasma, whole
processes document assay validity, accuracy, and precision, and blood to whole blood. Neither primary nor secondary stan-
should include external quality assessment, reference interval dards are assayed during routine patient sample testing, only
preparation and publication, and lot-to-lot validation.3 during calibration.
Preanalytical variables are addressed in Chapter 3, which Unfortunately, in hematology and hemostasis, in which
discusses blood specimen collection, and in Chapter 45, the analytes are often cell suspensions or enzymes, there are
which includes a section on coagulation specimen manage- just a handful of primary standards: cyanmethemoglobin,
ment. Postanalytical variables are discussed at the end of this fibrinogen, factor VIII, protein C, antithrombin, and von
chapter. Willebrand factor.7 For scores of analytes, the hematology and
The control of analytical variables begins with laboratory hemostasis laboratory scientist relies on calibrators. Calibrators
assay validation. for hematology may be preserved human blood cell suspen-
sions, sometimes supplemented with microlatex particles or
nucleated avian red blood cells (RBCs) as surrogates for hard-
VALIDATION OF A NEW OR
to-preserve human white blood cells (WBCs). In hemostasis,
MODIFIED ASSAY
calibrators may be frozen or lyophilized plasma from healthy
All new laboratory assays and all assay modifications require human donors. For most analytes it is impossible to prepare
validation.4 Validation is comprised of procedures to deter- weighed-in standards; instead, calibrators are assayed using
mine accuracy, specificity, precision, limits, and linearity.5 The reference methods (gold standards) at selected independent
results of these procedures are faithfully recorded and made expert laboratories. For instance, a vendor may prepare a 1000-L
available to on-site assessors upon request.6 lot of preserved human blood cell suspension, assay for the
desired analytes in house, and send aliquots to five laboratories
Accuracy that employ well-controlled reference instrumentation and
Accuracy is the measure of concurrence or difference between methods. The vendor obtains blood count results from all five,
an assay value and the theoretical true value of an analyte averages the results, compares them to the in-house values, and
(Figure 5-1). Some statisticians prefer to define accuracy as publishes the averages as the reference calibrator values. The
the extent of error between the assay result and the true vendor then distributes sealed aliquots to customer laborato-
value. Accuracy is easy to define but difficult to establish and ries with the calibrator values published in the accompanying
maintain. package inserts. Vendors often market calibrators in sets of
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 43

Gaussian, accurate, Gaussian, inaccurate,


and precise but precise

Dispersion
Frequency

Frequency
Target Target

Value Value

Gaussian, accurate, Gaussian, but inaccurate,


but imprecise and imprecise
Frequency

Frequency

Target Target

Value Value
Figure 5-1 The values generated by repeated assays of an analyte are graphed as a frequency distribution. Incremental values are plotted on the horizontal
(x) scale and number of times each value was obtained (frequency) on the vertical (y) scale. In this example, the values are normally distributed about their
mean (gaussian distribution). An accurate assay generates a mean that closely duplicates the reference target value. A precise assay generates small disper-
sion about the mean, whereas imprecision is reflected in a broad curve. The ideal assay is both accurate and precise.

three or five, spanning the range of assay linearity or the range Medical laboratory scientists may employ locally collected
of potential clinical results. fresh normal blood as a calibrator; however, the process for
As with secondary standards, vendors attempt to match validation and certification is laborious, so few attempt it. The
their calibrators as closely as possible to the physical properties selected specimens are assayed using reference equipment and
of the test sample. For instance, human preserved blood used methods, calibration values are assigned, and the new or modi-
to calibrate complete blood count analytes is prepared to fied assay is calibrated from these values. The Student t-test is
closely match the matrix of fresh anticoagulated patient blood often the statistic employed to match the means of the refer-
specimens, despite the need for preservatives, refrigeration, and ence and of the new assay. Often the reference equipment and
sealed packaging. Vendors submit themselves to rigorous cer- methods are provided by a nearby laboratory.
tification by governmental or voluntary standards agencies in
an effort to verify and maintain the validity of their products. Determination of Accuracy by Regression Analysis
The scientist assays the calibration material using the new If a series of five calibrators is used, results may be analyzed by
or modified assay and compares results with the vendors pub- the following regression equation:
lished results. When new results parallel published results
within a selected range, for example 10%, the results are y = a + bx
recorded and the assay is validated for accuracy. If they fail to
XY ( X )( Y ) X ( X )
2
Slope (b ) = n n 2
match, the new assay is modified or a new reference interval
and therapeutic range is prepared. Intercept (a ) = Y b ( X ) n
44 PART I Introduction to Hematology

130 130
125 r = 1.0 125 r = 0.89
Y intercept = 0.0 Y intercept = 0.0
120 120
115 115
New Assay 1

New Assay 3
110 110
105 105
Line of identity
100 100
95 95
90 90
85 85
80 80
80 85 90 95 100 105 110 115 120 125 130 80 85 90 95 100 105 110 115 120 125 130
Reference Reference

130 130
125 r = 1.0 125 r = 0.89
Y intercept = 5.0 Y intercept = 5.0
120 120
115 115
New Assay 2

110 New Assay 4 110


105 105
100 100
95 95
90 90
85 85
80 80
80 85 90 95 100 105 110 115 120 125 130 80 85 90 95 100 105 110 115 120 125 130
Reference Reference
Figure 5-2 Linear regression comparing four new assays with a reference method. Assay 1 is a perfect match. The y intercept of assay 2 is 5.0, which
illustrates a constant systematic error, or bias. The regression (r) value for assay 3 is 0.89, which illustrates a proportional systematic error. New assay 4 has
both bias and proportional error.

where x and y are the variables; a = intercept between the regres- systematic error, but must modify the published reference
sion line and the y-axis; b = slope of the regression line; n = interval.
number of values or elements; X = first score; Y = second score; Regression analysis gains sufficient power when 100 or
XY = sum of the product of first and second scores; X = sum more patient specimens are tested using both the new and
of first scores; Y = sum of second scores; X2 = sum of squared reference assay in place of or in addition to calibrators. Data
first scores. Perfect correlation generates a slope of 1 and a y may be entered into a spreadsheet program that offers an auto-
intercept of 0. Local policy establishes limits for slope and y matic regression equation.
intercept; for example, many laboratory directors reject a slope
of less than 0.9 or an intercept of more than 10% above or Precision
below zero (Figure 5-2). Unlike determination of accuracy, assessment of precision (dis-
Slope measures proportional systematic error; the higher the persion, reproducibility, variation, random error) is a simple
analyte value, the greater the deviation from the line of iden- validation effort, because it merely requires performing a series
tity. Proportional errors are caused by malfunctioning instru- of assays on a single sample or lot of reference material.8 Preci-
ment components or a failure of some part of the testing sion studies always assess both within-day and day-to-day varia-
process. The magnitude of the error increases with the concen- tion about the mean and are usually performed on three to five
tration or activity of the analyte. An assay with proportional calibration samples, although they may also be performed
error may be invalid. using a series of patient samples. To calculate within-day preci-
Intercept measures constant systematic error (or bias, in sion, the scientist assays a sample at least 20 consecutive times
laboratory vernacular), a constant difference between the new using one reagent batch and one instrument run. For day-to-
and reference assay regardless of assay result magnitude. A day precision, at least 10 runs on 10 consecutive days are
laboratory director may choose to adopt a new assay with required. The day-to-day precision study employs the same
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 45
sample source and instrument but separate aliquots. Day-to- 180
day precision accounts for the effects of different operators,
160 170.6
reagents, and environmental conditions such as temperature
140 146.2
and barometric pressure.
The collected data from within-day and day-to-day

Assayed, percent
120
sequences are reduced by formula to the mean and a measure
100 100.4
of dispersion such as standard deviation or, most often,
coefficient of variation in percent (CV%): 80

60
Mean ( x ) =
; 40
56.1
n
20 30.1
where (x) = the sum of the data values and n = the number 0
of data points collected 0 20 40 60 80 100 120 140 160 180 200
Computed, percent

Standard deviation ( s) =
(x x )
2 Figure 5-3 Determination of linearity. Prepare at least five dilutions of the
standard or calibrator. Dilutions must span the expected range of analyte
n 1
measurements. Compute the concentration of the analyte for each of the five
s
CV% = 100 dilutions. Plot the assayed values on the y scale and the computed concentra-
x tions on the x scale. Select the linear range by visual inspection, including
The CV% documents the degree of random error generated by the dilutions for which assayed values vary in a linear manner. In this
an assay, a function of assay stability. example, the limits of linearity are 56.1% to 146.2%. Assay results that fall
outside these limits are inaccurate.
CV% limits are established locally. For analytes based on
primary standards, the within-run CV% limit may be 5% or
less, and for hematology and hemostasis assays, 10% or less; within the linear range are valid; however, they must be mul-
however, the day-to-day run CV% limits may be as high as tiplied by the dilution. Laboratory scientists never report results
30%, depending upon the complexity of the assay. Although that fall below or above the linear limits, because accuracy is
accuracy, linearity, and analytical specificity are just as impor- compromised in the nonlinear regions of the assay. Lower
tant, medical laboratory scientists often equate the quality of limits are especially important when counting platelets or
an assay with its CV%. The best assay, of course, is one that assaying coagulation factors. For example, the difference
combines the lowest CV% with the greatest accuracy. between 1% and 3% factor VIII activity affects treatment
Precision for visual light microscopy leukocyte differential options and the potential for predicting coagulation factor
counts on stained blood films is immeasurably broad, particu- inhibitor formation. Likewise, the difference between a platelet
larly for low-frequency eosinophils and basophils.9 Most visual count of 10,000/mcL and 5000/mcL affects the decision to
differential counts are performed by reviewing 100 to 200 treat with platelet concentrate.
leukocytes. Although impractical, it would take differential
counts of 800 or more leukocytes to improve precision to Limits
measurable levels. Automated differential counts generated by Linearity studies are coupled with the lower limit of detection
profiling instruments, however, provide CV% levels of 5% or study. A zero calibrator, or blank, is assayed 20 times and
lower because these instruments count thousands of cells. the standard deviation is computed from the results. The lower
limit of detection is determined from the computed standard
Linearity deviation. The limit is three standard deviations above the
Linearity is the ability to generate results proportional to the mean of blank assays. This cutoff prevents false-positive results
calculated concentration or activity of the analyte. The labora- generated by low-end assay interference, commonly called
tory scientist dilutes a high-end calibrator or patient sample to noise. Limit assays are typically performed by the manufacturer
produce at least five dilutions spanning the full range of assay. or distributor and the results provided on the package insert;
The dilutions are then assayed. Computed and assayed results however, local policies often require that results of the manu-
for each dilution are paired and plotted on a linear graph, x facturers limit studies be confirmed.
scale and y scale, respectively. The line is inspected visually for
nonlinearity at the highest and lowest dilutions (Figure 5-3). Analytical Specificity
The acceptable range of linearity is established inboard based Analytical specificity is the ability of an assay to distinguish the
on the values at which linearity loss is evident. Although analyte of interest from anticipated interfering substances
formulas exist for computing the limits of linearity, visual within the sample matrix. The laboratory scientist spikes
inspection is the accepted practice. Nonlinear graphs may be identical samples with potential interfering substances and
transformed using semilog or log-log graphs when necessary. measures the effects of each upon the assay results. Specificity
Patient samples with results above the linear range must be is always determined by the manufacturer and need not be
diluted and reassayed. Results from diluted samples that fall confirmed at the local providers site unless there is suspicion
46 PART I Introduction to Hematology

TABLE 5-1 Categories of Laboratory Assay Approval TABLE 5-2 Example of a Lot-to-Lot Comparison
Assay Category Comment Old Lot New Lot
Sample Value Value % Difference
Food and Drug The local institution may use package insert
Administration data for linearity and specificity but must Low 7 6 14%
cleared establish accuracy and precision. Low middle 12 12
Analyte-specific Manufacturer may provide individual reagents Middle 20.5 19.4 5%
reagent but not in kit form, and may not provide High middle 31 27 20%
package insert validation data. Local High 48 48
institution must perform all validation steps. Old kit control 1 9 11 8%
Research use only Local institution must perform all validation Old kit control 2 22 24 8%
steps. Research use only assays are New kit control 1 10 10
intended for clinical trials, and carriers are New kit control 2 24 24
not required to pay.
Home brew Assays devised locally, all validation studies
are performed locally. optimistically no more than once a year. The new reagent lot
must arrive approximately a month before the laboratory runs
out of the old lot so that lot-to-lot comparisons may be completed
of interference from a particular substance not assayed by the and differences resolved, if necessary. The scientist uses control
manufacturer. Manufacturer specificity data are transferred or patient samples and prepares a range of analyte dilutions,
from the package insert to the local validation report. typically five, spanning the limits of linearity. If the reagent kits
provide controls, these are also included, and all are assayed
Levels of Laboratory Assay Approval using the old and new reagent lots. Results are charted as illus-
The U.S. Food and Drug Administration (FDA) categorizes trated in Table 5-2.
assays as cleared, analyte-specific reagent (ASR) assays, research Action limits vary by location, but many managers reject the
use only (RUO) assays, and home-brew assays. FDA-cleared new lot when more than one sample generates a variance
assays are approved for the detection of specific analytes and greater than 10% or when all variances are positive or negative.
should not be used for off-label applications. Details are given In the latter case, the new lot may be rejected or it may be
in Table 5-1. necessary to use the lot but develop a new reference interval
and therapeutic range.
Documentation and Reliability
Validation is recorded on standard forms. The Clinical Labora-
DEVELOPMENT OF THE REFERENCE
tory Standards Institute (CLSI) and David G. Rhoads Associates
INTERVAL AND THERAPEUTIC RANGE
(http://www.dgrhoads.com/files1/EE5SampleReports.pdf)
provide automated electronic forms. Validation records are Once an assay is validated, the laboratory scientist develops the
stored in prominent laboratory locations and made available reference interval (reference range, normal range).10 Most prac-
to laboratory assessors upon request. titioners tend to use the vernacular term normal range; however,
Precision and accuracy maintained over time provide assay reference interval is the preferred term. According to the
reliability. The recalibration interval may be once every 6 mathematical definitions, range encompasses all assay results
months or in accordance with operators manual recommenda- from largest to smallest, whereas interval is a statistic that
tions. Recalibration is necessary whenever reagent lots are trims outliers.
updated unless the laboratory can demonstrate that the report- To develop a reference interval the scientist carefully defines
able range is unchanged using lot-to-lot comparison. When the desired normal population and recruits representative
control results demonstrate a shift or consistently fall outside donors who meet the criteria to provide blood specimens. The
action limits, or when an instrument is repaired, the validation definition may, for example, exclude smokers, women taking
procedure is repeated. oral contraceptives, and people using over-the-counter or pre-
Regularly scheduled validity rechecks, lot-to-lot compari- scription medications. Donors may be paid. There should be
sons, instrument preventive maintenance, staff competence, an equal number of males and females, and the normal subjects
and scheduled performance of internal quality control and should match the institutions population demographics in
external quality assessment procedures assure continued reli- terms of age and race. When practical, large-volume blood
ability and enhance the value of a laboratory assay to the specimens are collected, aliquoted, and placed in long-term
patient and physician. storage. For instance, plasma aliquots for coagulation reference
interval development are stored at 70 C. It may be impracti-
cal to develop local reference intervals for infants, children, or
LOT-TO-LOT COMPARISONS
geriatric populations. In these cases the laboratory director may
Laboratory managers reach agreements with vendors to seques- choose to use published (textbook) intervals. In general,
ter kit and reagent lots, which ensures infrequent lot changes, although published normal values are available for educational
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 47

68.26%

95.46%
0.14% 0.14%
99.73%
-3s -2s -1s x +1s +2s +3s
Figure 5-4 Normal (gaussian) distribution. When the test values obtained for a given subject population are normally distributed, the mean is at the peak.
The segments of the population distribution representing 1, 2, and 3 standard deviations are illustrated. In developing the reference interval, laboratory
directors often use 2 standard deviations to establish the 95.5% confidence interval. This means that 95.46% of the test values from the normal (healthy)
population are included within 2 standard deviations. Consequently, 4.54%, or approximately 1 in 20 normal test results, fall outside the interval, half (2.27%)
above and half below.

and general discussion purposes, local laboratories must gener- limits at 2 standard deviations encompass 95.46% of normal
ate their own reference intervals to most closely match the results, known as the 95% confidence interval. This implies that
demographics of the area served by their institution. 4.54% of theoretically normal results fall outside the interval.
The minimum number of subject samples required to A standard deviation computed from a nongaussian distribu-
develop a reference interval may be determined using statistical tion may turn out to be too narrow to reflect the true reference
power computations; however, practical limitations prevail.11 interval and may thus encompass fewer than 95% of theoreti-
For a completely new assay with no currently established refer- cal normal values and generate a number of false positives.
ence interval, a minimum of 120 samples is necessary. In most Assays with high CV% values have high levels of random error
cases, however, the assay manufacturer provides a reference reflected in a broad curve; low CV% assays with tight disper-
interval on the package insert, and the local laboratory scientist sal have smaller random error and generate a narrow curve, as
need only assay 30 samples, 15 male and 15 female, to validate illustrated in Figure 5-1. The breadth of the curve may also
the manufacturers reference interval, a process called transfer- reflect biologic variation in values of the analyte.
ence. Likewise, the scientist may refer to published reference A few hematology and hemostasis assays are used to
intervals and, once they are locally validated, transfer them to monitor drug therapy. For instance, the international normal-
the institutions report form. ized ratio (INR) for prothrombin time is used to monitor the
It is assumed that the population sample subjects employed effects of oral Coumadin (warfarin) therapy, and the therapeu-
to generate reference intervals will produce frequency distribu- tic range is universally established at an INR of 2 to 3. On the
tions (in laboratory vernacular, histograms) that are normal bell- other hand, the therapeutic range for monitoring treatment
shaped (gaussian) curves (Figure 5-4). In a gaussian frequency with unfractionated heparin using the partial thromboplastin
distribution the mean is at the center and the dispersion about time (PTT) assay must be established locally by performing
the mean is identical in both directions. In many instances, regression of the PTT results in seconds against the results of
however, biologic frequency distributions are log-normal with the chromogenic anti-Xa heparin assay, whose therapeutic range
a tail on the high end. For example, it has been assumed for is established empirically as 0.3 to 0.7 international heparin
years that the visual reticulocyte percentage reference interval in units. The PTT therapeutic range is called the Brill-Edwards curve
adults is 0.5% to 1.5%; however, repeated analysis of normal and is described in Chapter 45.
populations in several locations has established it at 0.5% to If assay revalidation or lot-to-lot comparison reveals a sys-
2%, owing to a subset of normal subjects whose reticulocyte tematic change caused by reagent or kit modifications, a new
counts fall at the high end of the range. Scientists may choose reference interval (and therapeutic range, when applicable) is
to live with a log-normal distribution or they may transform it established. The laboratory director must warn the hospital
by replotting the curve using a semilog or log-log graphic staff of reference interval and therapeutic range changes,
display. The decision to transform may arise locally but eventu- because failure to observe new intervals and ranges may result
ally becomes adopted as a national practice standard. in diagnosis and treatment errors.
In a normal distribution, the mean ( x ) is computed by
dividing the sum of the observed values by the number of data
INTERNAL QUALITY CONTROL
points, n, as shown in the equation on page 45. The standard
deviation is calculated using the second formula in that equa- Controls
tion. A typical reference interval is computed as 2 standard Laboratory managers prepare, or more often purchase, assay
deviations and assumes that the distribution is normal. The controls. Although it may appear similar, a control is wholly
48 PART I Introduction to Hematology

distinct from a calibrator. Indeed, cautious laboratory directors single-run and long-term control variation are a function
may insist that controls be purchased from distributors differ- of assay dispersion or random error and reflect the CV% of
ent from those who supply their calibrators. As discussed in an assay.
the section Validation of a New or Modified Assay, calibrators Dr. James Westgard has established a series of internal
are used to adjust instrumentation or to develop a standard quality control rules that are routinely applied to long-term
curve. Calibrators are assayed by a reference method in expert deviations, called the Westgard rules.14 The rules were
laboratories and their assigned value is certified. Controls are developed for assays that employ primary standards, but a
used independently of the calibration process so that system- few Westgard rules that are the most useful in hematology
atic errors caused by deterioration of the calibrator or a change and hemostasis laboratories are provided in Table 5-4, along
in the analytical process can be detected through internal with the appropriate actions to be taken.15
quality control. This process is continuous and is called calibra-
tion verification.12 Compared with calibrators, control materials
are inexpensive and are comprised of the same matrix as Normal distribution
patient samples except for preservatives or freezing that provide +3s
a long shelf life. Controls provide known values and are
sampled directly alongside patient specimens to accomplish +2s
within-run assay validation. In nearly all instances, two con-
trols are required per test run, one in the normal range and +1s
the other in an expected abnormal range. For some assays

Mean
there is reason to select controls whose values are near the 0
interface of normal and abnormal. In institutions that perform
continuous runs, the controls should be run at least once per -1s
shift, for instance, at 7 AM, 3 PM, and 11 PM. In laboratories
where assay runs are discrete events, two controls are assayed -2s
with each run.
Control results must fall within predetermined dispersal -3s
limits, typically 2 standard deviations. Control manufacturers 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30
provide limits; however, local laboratory scientists must vali- Day
date and transfer manufacturer limits or establish their own, Control
usually by computing standard deviation from the first 20 Figure 5-5 Normal Levey-Jennings plot. Control results from 19 runs in
control assays. Whenever the result for a control is outside the 20 days all fall within the action limits established as 2 standard deviations
limits the run is rejected and the cause is found and corrected. (s). Results distribute evenly about the mean.
The steps for correction are listed in Table 5-3.
Control results are plotted on a Levey-Jennings chart that
displays each data point in comparison to the mean and limits Shift
(Figure 5-5).13 The Levey-Jennings chart assumes that the +3s
control results distribute in a gaussian manner and provide
limits at 1, 2, and 3 standard deviations about the mean. In +2s
addition to being analyzed for single-run errors, the data points
are examined for sequential errors over time (Figure 5-6). Both +1s
Mean

0
TABLE 5-3 Steps Used to Correct an Out-of-Control
Assay Run -1s

Step Description
-2s
1. Reassay When a limit of 2 standard deviations
is used, 5% of expected assay results -3s
fall above or below the limit. 0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30
2. Prepare new control Controls may deteriorate over time when Day
and reassay exposed to adverse temperatures or Control
subjected to conditions causing Figure 5-6 Levey-Jennings plot that illustrates a systematic error or shift.
evaporation. Control results from 21 runs in 22 days all fall within the action limits estab-
3. Prepare fresh reagents Reagents may have evaporated or lished as 2 standard deviations (s); however, the final 11 control results
and reassay become contaminated. are above the mean. When 10 consecutive control results fall on one side
4. Recalibrate instrument Instrument may require repair. of the mean, the assay has been affected by a systematic error (shift). The
operator troubleshoots and recalibrates the assay.
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 49

TABLE 5-4 Westgard Rules Employed in Hematology control samples but provides additional protection against
and Hemostasis shifts and trends.

12s or 22s A single control assay or two control assays are outside the
Delta Checks
2 standard deviation limit. Assay results are held until
The -check system compares a current analyte result with the
the error is identified using the steps in Table 5-3.
result from the most recent previous analysis for the same
Variations of this rule are 13s and 41s.
patient.17 Patient values remain relatively consistent over time
13s A single control assay is outside the 3 standard deviation
unless there is an intervention. A result that fails a check is
limit. Assay results are held until the error is identified
investigated for intervention or a profound change in the
using the steps in Table 5-3.
patients condition subsequent to the previous analysis. If there
R4s Two consecutive control values are more than 4 standard
is no ready explanation, the failed check may indicate an
deviations apart. Assay results are held until the error is
analytical error or mislabeled specimen. Results that fail a
identified using the steps in Table 5-3.
check are sequestered until the cause is found. Not all assays
Shift A series of at least 10 control values remain within the
are amenable to checking; only analytes known to show little
dispersal limits but are consistently above or below the
intraindividual variation are checked. These include MCV and
mean. Use of the assay is suspended until the cause is
RBC distribution width. In hemostasis, the prothrombin time
found; often it is an instrument calibration issue that has
and INR are checked. Action limits for checks are based on
introduced a constant systematic error (bias).
clinical impression and are assigned by hematology and hemo-
Trend A series of at least 10 control values changes in a
stasis laboratory directors in collaboration with the house and
consistent direction. Use of the assay is suspended until
laboratory staff. Computerization is essential, and checks are
the cause is found; often it is an instrument calibration
designed only to identify gross errors, not changes in random
issue that has introduced a systematic proportional error.
error, or shifts or trends. There is no regulatory requirement for
In hematology, shifts may be caused by deterioration of reagents, pump fittings, or checks.
light sources. Abrupt shifts may reflect a reagent or instrument fitting change.

EXTERNAL QUALITY ASSESSMENT


Moving Average ( X B) of the Red Blood External quality assessment further validates the accuracy of
Cell Indices hematology and hemostasis assays by comparing results from
In 1974, Dr. Brian Bull proposed a method of employing identical aliquots of samples distributed at regular intervals
patient RBC indices to monitor the stability of hematology among laboratories nationwide or worldwide. The aliquots are
analyzers, recognizing that the RBC indices mean cell volume often called survey or proficiency testing samples and include
(MCV), mean cell hemoglobin (MCH), and mean cell hemo- preserved human donor plasma and whole blood, stained
globin concentration (MCHC) remain constant on average blood and bone marrow smears, and photomicrographs of
despite individual patient variations.16 Each consecutive cells or tissues.
sequence of 20 patient RBC index assay results is collected and In most proficiency testing systems, target (true or refer-
treated by the moving average formula, which accumulates, ence) values for the test samples are established in-house by
smoothes, and trims data to reduce the effect of outliers. their manufacturer or distributor and are then further validated
Each trimmed 20-sample mean, X B, is plotted on a Levey- by preliminary distribution to a handful of expert laborato-
Jennings chart and tracked for trends and shifts using Westgard ries. Separate target values may be assigned for various assay
rules. The formula has been automated and embedded in the methods and instruments, as feasible.
circuitry of all hematology analyzers, which provide a Levey- Laboratories that participate in external quality assessment
Jennings graph for MCV, MCH, and MCHC. The moving are directed to manage the survey samples using the same
average concept has been generalized to WBC and platelet sample-handling principles as those employed for patient
counts and to some clinical chemistry analytes, albeit with specimenssurvey samples should not receive special atten-
moderate success. tion. Turnaround is swift, and results are sent electronically to
To begin, 500 consecutive samples are analyzed for the the provider.
mean MCV, MCH, and MCHC. A Levey-Jennings chart is pre- In addition to establishing a target value, agencies that
pared using 3% of the mean or one standard deviation as the administer surveys reduce the returned data to statistics, includ-
action limits, and subsequent data accumulation commences ing the mean, median, and standard deviation of all participant
in groups of 20. results. Provided the survey is large enough, the statistics may
The moving average method requires a computer to calcu- be computed individually for the various instruments and
late the averages, does not detect within-run errors, and is less assay methods. The statistics should match the predetermined
sensitive than the use of commercial controls in detecting sys- targets. If they do not, the agency troubleshoots the assay and
tematic shifts and trends. It works less well in institutions that assigns the most reliable statistics, usually the group mean and
assay samples from specialized populations, such as sickle cell standard deviations.
or oncology populations, for which the index values may The agency provides a report to each laboratory, illustrating
include a number of outliers. It does not replace the use of its result in comparison with the target value and appending a
50 PART I Introduction to Hematology

comment if the laboratory result exceeds the established limits, diagnosis notes, or the results of existing laboratory tests,
usually 2 standard deviations from the mean. If the specimen excluding the new assay. The new assay is then applied to
is a blood or bone marrow smear, a photomicrograph, or a samples from both the normal and disease groups to assess its
problem that requires a binary (positive/negative, yes/no) efficacy.
response, the local laboratory comment is compared with In a perfect world, the laboratory scientist sets the discrimi-
expert opinion and consensus. nation threshold at the 95% confidence interval limit (2 stan-
Although a certain level of error is tolerated, error rates that dard deviations) of the reference interval. When this threshold
exceed established limits result in corrective action or, in is used, the test will hopefully yield a positive result (e.g.,
extreme circumstances, loss of laboratory accreditation or elevated or reduced level) in every instance of disease and a
licensure. negative result (within the reference interval) in all normal
There are a number of external quality assessment agencies; control subjects. In reality, there is always overlap: a gray area
however, the College of American Pathologists (CAP) provides in which some positive test results are generated by normal
the largest survey systems, CAP accreditation, and CAP Surveys/ samples (false positives) and some negative results are generated
EXCEL proficiency testing. Survey packages are provided for by samples from patients with disease (false negatives). False
laboratories offering all levels of service. CAP is a nongovern- positives cause unnecessary anxiety, follow-up expense, and
mental agency; however, survey participation is necessary to erroneous diagnostic leadsworrisome, expensive, and time
meet the accreditation requirements of the Joint Commission consuming, but not fatal. False negatives fail to detect the
(formerly the Joint Commission on Accreditation of Health- disease and may delay treatment, which is potentially life-
care Organizations) and to qualify for Medicare reimburse- threatening. The laboratory scientist employs clinical efficacy
ment. The North American Specialized Coagulation Laboratory computations to establish laboratory assay efficacy and
Association provides survey systems for specialty coagulation minimize both false positives and false negatives (Table 5-5).
laboratories in the United States and Canada, and is affiliated Clinical efficacy testing includes determination of diagnostic
with the ECAT (external quality control of diagnostic assays sensitivity and specificity, and positive and negative predictive
and tests) Foundation External Quality Assessment Program value, as well as receiver operating characteristic analysis.
of The Netherlands, which provides survey materials through- To start a clinical efficacy study the scientist selects normal
out Europe. Many state health agencies provide proficiency control samples and samples from people proven to have the
testing surveys, requiring laboratories to participate as a condi- disease or condition addressed by the assay. To make the dis-
tion of licensure. cussion easy, assume that 50 samples of each are chosen. All
are assayed, and the results turn out as shown in Table 5-6.
The scientist next computes clinical sensitivity and speci
MEASUREMENT OF CLINICAL EFFICACY
ficity and positive and negative predictive value as shown in
Since 1940 and before, surgeons have used the bleeding time Table 5-7.
test to predict the risk of intraoperative hemorrhage. The labo-
ratory scientist or phlebotomist activates an automated lancet
TABLE 5-5 Clinical Efficacy Definitions and Binary
to make a 5-mm long, 1-mm deep incision in the volar surface
Display
of the forearm and, using a filter paper to soak up the blood,
times the interval from the initial incision to bleeding cessa- True positive Assay correctly identifies a disease or condition in
tion, normally 2 to 9 minutes. The test is simple and logical, those who have it.
and for over 50 years experts have claimed that if the incision False positive Assay incorrectly identifies disease when none is
bleeds for longer than 9 minutes, there is a risk of surgical present.
bleeding. In the 1990s clinical researchers compared normal True negative Assay correctly excludes a disease or condition in
and prolonged bleeding times with instances of intraoperative those without it.
bleeding and found to their surprise that prolonged bleeding False negative Assay incorrectly excludes disease when it is present.
time predicted only 50% of intraoperative bleeds.18,19 The other
50% occurred despite a normal bleeding time. Thus the posi- Normal Disease or Condition
tive predictive value of the bleeding time for intraoperative (Control Sample) (Patient Sample)
bleeding was 50%, the same as the probability of obtaining
Assay is negative True negative False negative
heads (or tails) in tossing a coin. Today the bleeding time test
Assay is positive False positive True positive
is widely agreed to have no clinical relevance and is obsolete.
Like the bleeding time test, many time-honored hematology
and hemostasis assays gain credibility on the basis of expert
TABLE 5-6 Clinical Efficacy Study
opinion. Now, however, besides being valid, accurate, linear,
and precise, a new or modified assay must be clinically effec- Normal Disease or Condition
tive.20 To compute clinical efficacy, the scientist uses a series of (Control Sample) (Patient Sample)
samples from normal healthy subjects, called controls, and Assay is negative True negative: 45 False negative: 5
patients who conclusively possess a disease or condition. The Assay is positive False positive: 5 True positive: 45
patients diagnosis is based on clinical outcomes, discharge
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 51

TABLE 5-7 Clinical Efficacy Computations


Statistic Definition Formula Example
Sensitivity Proportion with the disease who have a Sensitivity (%) = TP/(TP + FN) 100 45/(45 + 5) 100 = 90%
positive test result
Distinguish clinical sensitivity from analytical sensitivity. Analytical sensitivity is a measure of the smallest increment of the analyte that can be
distinguished by the assay.
Specificity Proportion without the disease who have a Specificity (%) = TN/(TN + FP) 100 45/(45 + 5) 100 = 90%
negative test result
Distinguish clinical specificity from analytical specificity. Analytical specificity is the ability of the assay to distinguish the analyte from interfering substances.
Positive predictive value (PPV) Proportion with a disease who have a PPV (%) = TP/(TP + FP) 100 45/(45 + 5) 100 = 90%
positive test result compared with all
subjects who have a positive test result
The positive predictive value predicts the probability that an individual with a positive assay result has the disease or condition.
Negative predictive value (NPV) Proportion without a disease who have a NPV (%) = TN/(TN + FN) 100 45/(45 + 5) 100 = 90%
negative test result compared with all
subjects who have a negative test result
The negative predictive value predicts the probability that an individual with a negative assay result does not have the disease or condition.

FN, False negative; FP, false positive; TN, true negative; TP, true positive.

For all assays, as sensitivity rises, specificity decreases. Assays 2 standard deviation limit of the reference interval is used as
with high sensitivity and low specificity make effective screen- the threshold for discriminating a positive from a negative test
ing tests, although they produce a number of false positives. result. Often the true threshold varies from the 2 standard
For instance, if the condition being studied has a prevalence of deviation limit. Using ROC analysis, the discrimination thresh-
0.0001 (1 in 10,000) and the false-positive rate is 1%, the assay old is adjusted by increments of 1, and the true-positive and
will produce 99 false-positive results for every true-positive false-positive rates are recomputed for each new threshold
result. Clearly such a test is useful only when the consequence level. The threshold that is selected is the one that provides the
of a false-positive result is minimal. largest true-positive and smallest false-positive rate (Figure
Likewise, as specificity rises, sensitivity goes down. Assays 5-7). A line graph is generated plotting true positives on the
with high specificity make effective confirmatory assays when y-axis and false positives on the x-axis. The overall efficacy of
used in follow-up to positive results on screening assays. High- the assay is assessed by measuring the area under the curve. If
specificity assays produce a number of false negatives and the area under the curve is 0.5, the curve is at the line of identity
should not be used as initial screens. A positive result on both between false and true positives and provides no discrimina-
a screening assay and a confirmatory assay provides a definitive tion. Most agree that a clinically useful assay should have an
conclusion. A positive screening result followed by a negative area under the curve of 0.85 or higher.
confirmatory test result generates a search for alternative
diagnoses.
ASSAY FEASIBILITY
Laboratory assays are most effective when used for patients
with high clinical pretest probability. In such instances, the Most laboratory managers and directors review assay feasibility
prevalence of the condition is high enough to mitigate the before launching complex validation, efficacy, and quality
effects of false positives and false negatives. When a physician control initiatives. Feasibility study includes a review of assay
orders hemostasis testing for patients who are experiencing throughput (number of assays per unit time), cost per test,
easy bruising, there is a high pretest probability, which raises turnaround time, and the technical skill required to perform
the tests clinical efficacy. Ordering hemostasis tests routinely the assay. To select a new instrument, the manager reviews
for healthy individuals prior to elective surgery presumes issues of operator safety, footprint, overhead, compatibility
a low pretest probability and reduces the efficacy of the test with laboratory utilities and information system, the need for
profile. middleware, frequency and duration of breakdowns, and dis-
tributor support and service.

RECEIVER OPERATING
CHARACTERISTIC CURVE LABORATORY STAFF COMPETENCE
A receiver operating characteristic (ROC) curve or ROC analysis Staff integrity and professional staff competence are the
is a further refinement of clinical efficacy testing that may be keys to assay reliability. In the United States, 12 states have
employed when the assay being validated generates a continu- medical laboratory personnel licensure laws. In these states,
ous variable.21 In clinical efficacy testing as described earlier, the only licensed laboratory professionals may be employed in
52 PART I Introduction to Hematology

1.00
0.90
Threshold FP rate TP rate 0.80
70% 0.02 0.51
71% 0.05 0.62 0.70

True-positive rate
72% 0.10 0.80
73% 0.15 0.85 0.60
74% 0.30 0.89 0.50
75% 0.35 0.91
76% 0.38 0.93 0.40
77% 0.44 0.96 AUC ~0.85
78% 0.47 0.96 0.30
79% 0.51 0.98 0.20
80% 0.57 0.98
0.10
0.00
0.00 0.20 0.40 0.60 0.80 1.00
A False-positive rate

1.00
0.90
Cutoff FP rate TP rate 0.80
70% 0.02 0.31
71% 0.05 0.45 0.70
True-positive rate

72% 0.10 0.55


73% 0.15 0.65 0.60
74% 0.30 0.79 0.50
75% 0.35 0.81
76% 0.42 0.83 0.40
77% 0.44 0.86 AUC ~0.70
78% 0.47 0.86 0.30
79% 0.51 0.89 0.20
80% 0.57 0.90
0.10
0.00
0.00 0.20 0.40 0.60 0.80 1.00
B False-positive rate

1.00
0.90
Cutoff FP rate TP rate 0.80
70% 0.05 0.30
71% 0.10 0.35 0.70
True-positive rate

72% 0.15 0.40


73% 0.20 0.45 0.60
74% 0.25 0.50 0.50
75% 0.30 0.55
76% 0.35 0.60 0.40
77% 0.40 0.65 AUC ~0.50
78% 0.45 0.70 0.30
79% 0.50 0.75 0.20
80% 0.55 0.80
0.10
0.00
0.00 0.20 0.40 0.60 0.80 1.00
C False-positive rate
Figure 5-7 A, Receiver operating characteristic curve. The false-positive and true-positive rates for each discrimination threshold from 70% to 80% are
computed and graphed as paired variables on a linear scale, false-positive rate on the horizontal (x) scale and true-positive rate on the vertical (y) scale. The
assay is acceptable, with an area under the curve (AUC) of 0.85, and 73% is the threshold that produces the most desirable false-positive and true-positive
rates. B, This assay is unacceptable, with an AUC of 0.70. It is difficult to find the threshold that produces the most desirable false-positive and true-positive
rates. C, This assay, with an AUC of 0.50, has no clinical efficacy. FP, False positive; TP, true positive.
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 53

medical center or reference laboratories. In nonlicensure states the United States, professional levels are defined as the asso
conscientious laboratory directors employ only nationally ciate (2-year) degree level, or medical laboratory technician;
certified professionals. Certification is available from the bachelor (4-year) degree level, or medical laboratory scientist;
American Society for Clinical Pathology Board of Certification and the level of advanced degree, masters degree, or doctorate
in Chicago, Illinois. Studies of outcomes and laboratory in clinical laboratory science and related sciences. Many col-
errors demonstrate that laboratories that employ only licensed leges and universities offer articulation programs that enable
or certified professionals produce the most reliable assay professional personnel to advance their education and res
results.22,23 ponsibility levels. Several of these institutionsfor example,
Competent laboratory staff members continuously watch the University of Cincinnati, University of North Dakota,
for and document errors by inspecting the results of internal Weber State University, and University of Medicine and Den-
validation and quality control programs and external quality tistry in New Jerseyprovide distance learning opportunities.
assessment. Error is inevitable, and such incidents should be Enlightened employers encourage personnel to participate in
used for quality improvement and instruction. When error is advanced educational programs, and many provide resources
associated with liability, the opportunity for improvement is for this purpose. Education contributes to quality laboratory
often lost to obfuscation. The analysis of error without blame services.
may be consistently practiced in an effort to improve the
quality of laboratory service.
QUALITY ASSURANCE PLAN:
PREANALYTICAL AND POSTANALYTICAL
Proficiency Systems
Laboratory managers and directors assess and document pro- In addition to keeping analytical quality control records, U.S.
fessional staff skills using proficiency systems. The hematology agencies require laboratory directors to maintain records of
laboratory manager may, for instance, maintain a collection of preanalytical and postanalytical quality assurance and quality
normal and abnormal blood films, case studies, or laboratory improvement efforts.24 Although not exhaustive, Table 5-8 lists
assay reports that staff are required to examine at regular inter- and characterizes a number of examples of preanalytical quality
vals. Personnel who fail to reproduce the target values on efforts, and Table 5-9 provides a review of postanalytical com-
examination of the blood film are provided remedial instruc- ponents. All quality assurance plans include objectives, sources
tion. The proficiency set may also be used to assess applicants of authority, scope of services, an activity calendar, corrective
for laboratory positions. Proficiency testing systems are avail- action, periodic evaluation, standard protocol, personnel
able from external quality assessment agencies, and proficiency involvement, and methods of communication.25
reports are made accessible to laboratory assessors. CAP Q-PROBES is a subscription service that provides a
model quality assurance program. Experts in quality assurance
Continuing Education continuously refine the consensus of appropriate indicators of
The American Society for Clinical Pathology Board of Certifica- laboratory medicine quality. The Q-PROBES program searches
tion and state medical licensure boards require professional for events that provide improvement opportunities.
personnel to participate in and document continuing educa-
tion for periodic recertification or relicensure. Continuing educa-
AGENCIES THAT ADDRESS HEMATOLOGY
tion is delivered in the form of journal articles, case studies,
AND HEMOSTASIS QUALITY
distance learning seminars, and professional meetings. Medical
centers offer periodic internal continuing education opportuni- The following are agencies that are concerned with quality
ties (in-service education) in the form of grand rounds, lectures, assurance in hematology and hemostasis laboratory testing:
seminars, and participative educational events. Presentation Clinical and Laboratory Standards Institute (CLSI) (http://
and discussion of local cases is particularly effective. Continu- www.clsi.org), 940 West Valley Road, Suite 1400, Wayne,
ing education maintains the critical skills of laboratory scien- PA 19087. Produces guidelines and standards for labora-
tists and provides opportunities to learn about new clinical and tory practice. Hemostasis documents include H21-A5,
technical approaches. The Colorado Association for Continu- H30-A2, and H47H58. Hematology standards include
ing Medical Laboratory Education (http://www.cacmle.org), H02-A4, H07-A3, and H22H46. Clinical efficacy method
the American Society for Clinical Laboratory Science (http:// evaluation, mostly EP suffix standards; quality assurance
www.ascls.org), the American Society for Clinical Pathology and quality management systems, mostly GP suffix stan-
(http://www.ascp.org), the American Society of Hematology dards are available.
(http://www.hematology.org), the National Hemophilia Foun- Center for Medicare and Medicaid Services (CMS) (http://
dation (http://www.hemophilia.org), CLOT-ED (http://www. www.cms.hhs.gov), 7500 Security Boulevard, Baltimore,
clot-ed.com), and the Fritsma Factor (http://www.fritsmafactor. MD 21244. Administers the laws and rules developed from
com) are examples of scores of organizations that direct their the Clinical Laboratory Improvement Amendments of 1988.
activities toward quality continuing education in hematology Establishes Current Procedural Terminology (CPT) codes,
and hemostasis. reimbursement rules, and test complexity.
The medical laboratory science profession stratifies pro College of American Pathologists (CAP) (http://www.cap.
fessional staff responsibilities by educational preparation. In org/apps/cap.portal), 325 Waukegan Road, Northfield, IL
54 PART I Introduction to Hematology

TABLE 5-8 Preanalytical Quality Assurance Components and the Laboratorys Responsibility
Preanalytical Component Laboratory Staff Responsibility
Test orders Conduct continuous utilization reviews to ensure that physician laboratory orders are comprehensive and appropriate
to patient condition. Inform physician about laboratory test availability and ways to avoid unnecessary orders.
Reduce unnecessary repeat testing.
Test request forms Are requisition forms legible? Can you confirm patient identity? Are physician orders promptly and correctly interpreted
and transcribed?
Is adequate diagnostic, treatment, and patient preparation information provided to assist the laboratory in appropriately
testing and interpreting results?
Stat orders and timeliness Do turnaround time expectations match clinical necessity and ensure that stat orders are reserved for medical
emergencies?
Specimen collection Is the patient correctly identified, prepared, and available for specimen collection? Is fasting and therapy status
appropriate for laboratory testing?
Is the tourniquet correctly applied and released at the right time? Are venipuncture sites appropriately cleansed?
Are timed specimens collected at the specified intervals? Are specimen tubes collected in the specified order? Are
additive tubes properly mixed? Are specimen tubes labeled correctly?
Specimen transport Are specimens delivered intact, sealed, and in a timely manner? Are they maintained at the correct temperature?
Specimen management Are specimens centrifuged correctly? Are tests begun within specified time frames? Are samples stored properly? Are
coagulation specimens platelet-poor when specified?

TABLE 5-9 Postanalytical Quality Assurance Components and the Laboratorys Responsibility
Postanalytical Component Laboratory Staff Responsibility
Publication of reports Are results accurately transcribed into the information system? Are they reviewed for errors by additional laboratory
staff? If autoverification is in effect, are the correct parameters employed?
Do reports provide reference intervals? Do they flag abnormal results? Are result narratives appended when necessary?
Does the laboratory staff conduct in-service education to support test result interpretation?
Are critical values provided to nursing and physician staff? Are verbal reports confirmed with feedback? Are anomalous
findings resolved?
Timeliness Are turnaround times recorded and analyzed? Are laboratory reports being posted to patient charts in a timely fashion?
Patient satisfaction Does the institution include laboratory care in patient surveys? Was specimen collection explained to the patient?

60093. Laboratory accreditation, proficiency testing, and Joint Commission (http://www.jointcommission.org), One
quality assurance programs; laboratory education, reference Renaissance Boulevard, Oakbrook Terrace, IL 60181.
resources, and e-lab solutions. Accreditation and certification programs.

SUMMARY
Each new assay or assay modification must be validated for accu- Internal quality control is accomplished by assaying controls with
racy, precision, linearity, specificity, and lower limit of detection each test run. Control results are compared with action limits,
ability. In the hematology and hemostasis laboratory, accuracy usually the mean of the control assay 2 standard deviations. If
validation usually requires a series of calibrators, although it may the control value is outside the limits, use of the assay is sus-
be accomplished by using a number of patient specimens and pended and the scientist begins troubleshooting. Control results
comparing results with those obtained using a reference method. are plotted on Levey-Jennings charts and examined for drift and
In all cases, accuracy is established using the Student t-test and trends. Internal quality control is enhanced through the use of the
linear regression. moving average algorithm.
Precision is established by using repeated within-day and day-to- All conscientious laboratory directors subscribe to an external
day assays, then computing the mean, standard deviation, and quality assessment system, also known as proficiency testing or
coefficient of variation of the results. proficiency surveys. External quality assessment enables the
Assay linearity, specificity, and lower limit of detection are usually director to compare selected assay results with other laboratory
provided by the vendor; however, many laboratory managers results, nationally and internationally, as a further check of accu-
require that these parameters be revalidated locally. racy. Maintaining a good external quality assessment record is
CHAPTER 5 Quality Assurance in Hematology and Hemostasis Testing 55

essential to laboratory accreditation. Most U.S. states require individual proficiency tests that are correlated with in-service edu-
external quality assessment for laboratory licensure. cation. Staff are encouraged to participate in continuing education
All laboratory assays are analyzed for clinical efficacy, sensitivity, activities and in-house discussion of cases. Quality laboratories
and specificity, including their true-positive and true-negative provide resources for staff to pursue higher education.
rates, and positive and negative predictive values. Highly sensitive The laboratory director maintains a protocol for assessing and
assays may be used for population screening, but may lack good improving upon preanalytical and postanalytical variables and
discrimination. Specific assays may be used to confirm a condi- finds means to communicate enhancements to other members of
tion, but generate a number of false negatives. Clinical efficacy the health care team.
computations expand to include receiver operating characteristic
curve analysis. Now that you have completed this chapter, go back and
Thoughtful laboratory managers hire only certified or licensed read again the case study at the beginning and respond
medical laboratory scientists and technicians and provide regular to the questions presented.

R E V I E W Q UESTIONS
1. You validate a new assay using linear regression to compare a. Cleared
assay calibrator results with the distributors published b. Home brew
calibrator results. The slope is 0.95 and y intercept is c. Research use only
+10%. What type of error is present? d. Analyte-specific reagent
a. No error
b. Random error 6. A laboratory scientist measures prothrombin time for
c. Constant systematic error plasma aliquots from 15 normal males and 15 normal
d. Proportional systematic error females. She computes the mean and 95.5% confidence
interval and notes that they duplicate the manufacturers
2. The acceptable hemoglobin control value range is 13 statistics within 5%. This procedure is known as:
0.4g/dL. The control is assayed five times and produces a. Confirming linearity.
the following five results: b. Setting the reference interval.
c. Determining the therapeutic range.
12.0g/dL 12.3g/dL 12.0g/dL 12.2g/dL 12.1g/dL
d. Establishing the reference interval by transference.
These results are:
a. Accurate but not precise. 7. You purchase a preserved whole blood specimen from
b. Precise but not accurate. a distributor who provides the mean values for several
c. Both accurate and precise. complete blood count analytes. What is this specimen
d. Neither accurate nor precise. called?
a. Normal specimen
3. A WBC count control has a mean value of 6000/mcL and b. Calibrator
a standard deviation of 300/mcL. What is the 95.5% con- c. Control
fidence interval? d. Blank
a. 3000 to 9000/mcL
b. 5400 to 6600/mcL 8. You perform a clinical efficacy test and get the following
c. 5500 to 6500/mcL results:
d. 5700 to 6300/mcL
Disease or
4. The ability of an assay to distinguish the targeted analyte Normal (Control Condition
from interfering substances within the sample matrix is Sample) (Patient Sample)
called:
Assay is negative 40 5
a. Analytical specificity.
b. Analytical sensitivity. Assay is positive 10 45
c. Clinical specificity.
d. Clinical sensitivity. What is the number of false-negative results?
a. 40
5. The laboratory purchases reagents from a manufacturer b. 10
and develops an assay using standard references. What c. 5
FDA category is this assay? d. 45
56 PART I Introduction to Hematology

9. What agency provides external quality assurance (profi- 11. Regular review of blood specimen collection quality is an
ciency) surveys and laboratory accreditation? example of:
a. College of American Pathologists (CAP) a. Preanalytical quality assurance.
b. Clinical Laboratory Improvement Advisory b. Analytical quality control.
Committee (CLIAC) c. Postanalytical quality assurance.
c. Center for Medicare and Medicaid Services (CMS) d. External quality assurance.
d. Joint Commission
12. Review of laboratory report integrity is an example of:
10. What agency provides continuing medical laboratory a. Preanalytical quality assurance.
education? b. Analytical quality control.
a. Clinical Laboratory Improvement Advisory c. Postanalytical quality assurance.
Committee (CLIAC) d. External quality assurance.
b. Center for Medicare and Medicaid Services (CMS)
c. Colorado Association for Continuing Medical
Laboratory Education (CACMLE)
d. College of American Pathologists (CAP)

REFERENCES
1. Westgard JO: Assuring the right quality right: good laboratory 13. Levey S, Jennings ER: The use of control charts in the clinical
practices for verifying the attainment of the intended quality of test laboratory. Am J Clin Pathol 20:10591066, 1950.
results, Madison, Wisc, 2008, Westgard QC. 14. Westgard JO, Quam EF, Barry PL, et al: Basic QC practices,
2. Clinical and Laboratory Standards Institute (CLSI): Continu- ed 2, Madison, Wisc, 2002, Westgard QC.
ous quality improvement: integrating five key quality system com- 15. Westgard JO, Barry PL, Hunt MR, et al: A multi-rule Shewhart
ponents; approved guideline, ed 2, CLSI document GP22-A2. chart for quality control in clinical chemistry. Clin Chem
Wayne, Pa, 2004, CLSI. 27:493501, 1981.
3. Clinical and Laboratory Standards Institute (CLSI): Procedures 16. Bull BS, Elashoff RM, Heilbron DC, et al: A study of various
for the collection of diagnostic blood specimens by venipuncture; estimators for the derivation of quality control procedures
approved standard, ed 6, CLSI document H3-A6. Wayne, Pa, from patient erythrocyte indices. Am J Clin Pathol 61:473
2004, CLSI. 481, 1974.
4. Westgard JO: Basic method validation, ed 3, Madison, Wisc, 17. Rhoads DG: Lab statisticsfun and easy, Kennett Square, Pa,
2008, Westgard QC. 2009, David G Rhoads Associates, Inc.
5. Clinical and Laboratory Standards Institute (CLSI): Method 18. Lind SE: The bleeding time does not predict surgical bleeding.
comparison and bias estimation using patient samples; approved Blood 77:25472552, 1991.
guideline, ed 2, CLSI document EP9-A2. Wayne, Pa, 2002, 19. Gewirtz AS, Miller ML, Keys TF: The clinical usefulness of the
CLSI. preoperative bleeding time. Arch Pathol Lab Med 120:353
6. McGlasson D, Plaut D, Shearer C: Statistics for the hemostasis 356, 1996.
laboratory, Bethesda, Md, 2006, ASCLS Press. 20. Fardy JM: Evaluation of diagnostic tests. Methods Mol Biol
7. Calibration and calibration verification. Clinical Laboratory 473:127136, 2009.
Improvement Amendments (CLIA) Brochure No. 3. Available 21. Sreide K: Receiver-operating characteristic curve analysis
at: http://www.phppo.cdc.gov/CLIA/regs.toc.asp. Accessed in diagnostic, prognostic and predictive biomarker research.
November 11, 2007. J Clin Pathol 62:15, 2009.
8. Clinical and Laboratory Standards Institute (CLSI): Laboratory 22. Novis DA: Detecting and preventing the occurrence of errors
instrument implementation, verification, and maintenance; in the practices of laboratory medicine and anatomic pathol-
approved guideline. CLSI document GP31-A, Wayne, Pa, 2009, ogy: 15 years experience with the College of American
CLSI. Pathologists Q-PROBES and Q-TRACKS programs. Clin Lab
9. Koepke JA, Dotson MA, Shifman MA: A critical evaluation of Med 24:965978, 2004.
the manual/visual differential leukocyte counting method. 23. Winkelman JW, Mennemeyer ST Using patient outcomes to
Blood Cells 11:173186, 1985. screen for clinical laboratory errors. Clin Lab Manage Rev.
10. Clinical and Laboratory Standards Institute (CLSI): Defining, 10:134136, 139142, 1996.
establishing and verifying reference intervals in the clinical labora- 24. Westgard JO, Ehrmeyer SS, Darcy TP: CLIA final rule for quality
tory; approved guideline, ed 3, CLSI document C28-A3. Wayne, systems, quality assessment issues and answers, Madison, Wisc,
Pa, 2004, CLSI. 2004, Westgard QC.
11. Cembrowski GS, Martindale RA: Quality control and statis- 25. Howanitz PJ, Hoffman GG, Schifman RB, et al: A nationwide
tics. In Bishop ML, Fody EP, Schoeff LE, editors: Clinical chem- quality assurance program can describe standards for the
istry: principles, procedures, correlations, ed 5, Philadelphia: practice of pathology and laboratory medicine. Qual Assur
2004 Lippincott Williams & Wilkins, chap 3, pp 48-89. Health Care 3:245256, 1992.
12. Bachner P: Quality assurance in hematology. In Howanitz JF,
Howanitz JH, editors: Laboratory quality assurance, New York,
1987, McGraw-Hill, pp 214243.
PART II Hematopoiesis

Cellular Structure and Function


Keila B. Poulsen
6
OUTLINE OBJECTIVES
Cell Organization After completion of this chapter, the reader will be able to:
Cell Membrane
Membrane Proteins 1. Describe the general function and chemical compo- 5. Correlate the cytoplasmic structures to the activities
Membrane Carbohydrates sition of cellular membranes. of the cell.
Nucleus 2. List and describe the components of the nucleus, 6. Describe the function of the hematopoietic inductive
Chromatin including staining qualities visible by light microenvironment.
Nuclear Envelope microscopy. 7. Associate stages in the cell cycle with activities of
Nucleoli 3. Correlate the nuclear structures to the activities of the cell.
Cytoplasm the cell. 8. Correlate regulatory proteins with the stage of the
Golgi Complex 4. Name and describe the ultrastructural appearance cell cycle.
Endoplasmic Reticulum of the cytoplasmic organelles found in the cell 9. Discuss the function of checkpoints in the cell cycle
Ribosomes
and staining qualities by light microscopy, if and where in the cycle they occur.
Mitochondria
appropriate. 10. Differentiate between apoptosis and necrosis.
Lysosomes
Microfilaments
Microtubules
Centrioles
Hematopoietic
Microenvironment
Cell Cycle
Regulation of the Cell
Cycle
Apoptosis

T
he numerous multichannel instruments available to and contain the components necessary to perform and per-
assist in the clinical diagnostic process have revolution- petuate these functions. Regardless of shape, size, or function,
ized the study of hematology. The technologies of light most cells have three basic parts: unit membranes, the cyto-
scatter, electrical impedance, and conductivity have added plasm, and the nucleus. Each of these basic parts has compo-
parameters and scatter plots whose significance is yet to be fully nents or subdivisions that assist in their varied functions. Table
realized and clinically applied, but morphologic examination 6-1 summarizes the cellular components and functions, which
of the peripheral blood film by light microscopy remains the are explained in more detail later.
hallmark for clinical evaluation of patients with hematologic
abnormalities. The study of cells under the microscope was
CELL MEMBRANE
greatly enhanced when Paul Ehrlich (1854-1915) developed
staining techniques to better differentiate the various normal The cell membrane serves as a semipermeable outer boundary
and abnormal cells present in human blood. The development separating the cellular components from their surrounding
of the electron microscope revolutionized the ability to study environment. The cell membrane serves three basic functions:
and understand the internal components of the cell.1 (1) it restricts and facilitates the interchange of substances
with the environment by selective permeability, endocytosis,
exocytosis, and locomotion; (2) it detects hormonal signals
CELL ORGANIZATION
facilitating cell-to-cell recognition; and (3) it is the location of
Cells are structural units that constitute living organisms surface markers for cell identity.2 Monoclonal antibodies are
(Figures 6-1 and 6-2). Many cells have specialized functions used to identify a cells surface markers. The nomenclature

57
58 PART II Hematopoiesis

Microfilaments Glycogen
aggregates

Golgi complex
Nuclear envelope
Chromatin Centriole
Nuclear pore
Rough
endoplasmic
reticulum
Microtubule
Perinucleolar
chromatin Vacuole

Nucleolus Mitochondria

Euchromatin

Heterochromatin Lysosome
Smooth
endoplasmic
reticulum
Free ribosomes

Figure 6-1 Cell organization.

Nuclear pore

Golgi body
Nucleus

Nucleolus

Lysosomes

Mitochondria Rough endoplastic reticulum


Figure 6-2 Electron micrograph with labeled organelles. (From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
CHAPTER 6 Cellular Structure and Function 59

TABLE 6-1 Summary of Cellular Components and Functions


Organelle Location Appearance and Size Function Comments
Membranes: Outer boundary of cell, Usually a lipid bilayer consisting Separates various cellular Membrane must be resilient
plasma, nuclear, nucleus, endoplasmic of proteins, cholesterol, components; facilitates and flexible
mitochondrial, reticulum, mitochondria, phospholipids, and and restricts cellular
endoplasmic and other organelles polysaccharides; membrane exchange of substances
reticulum thickness varies with cell or
organelle
Nucleus Within cell Usually round or oval but varies Control center of cell and Governs cellular activity and
depending on cell; varies in contains genetic transmits information for
size; composed of DNA blueprint cellular control
Nucleolus Within nucleus Usually round or irregular in shape; Site of synthesis and Appearance varies with activity
2-4m in size; composed of procession of ribosomal of cells; larger when cell is
RNA; there may be 1-4 within RNA actively involved in protein
nucleus synthesis
Golgi body Next to nucleus System of stacked, Involved in modifying Well developed in cells with
membrane-bound, flattened and packaging large secretion
sacs; horseshoe shaped; macromolecules for responsibilities
varies in size secretion
Endoplasmic Randomly distributed Membrane-lined tubules that Stores and transports Two types: smooth with no
reticulum throughout cytoplasm branch and connect to nucleus fluids and chemicals ribosomes; rough with
and plasma membrane ribosomes on surface
Ribosomes Free in cytoplasm; outer Small granule (100-300); Site of production of Large proteins are synthesized
surface of rough composed of protein and proteins, such as from polyribosomes (chains
endoplasmic reticulum nucleic acid enzymes and blood of ribosomes)
proteins
Mitochondria Randomly distributed in Round or oval structures; 3-14nm Cells powerhouse; make Active cells have more present
cytoplasm in length, 2-10nm in width; adenosine triphosphate, than do inactive cells
membrane has 2 layers; inner energy source for cell
layer has folds called cristae
Lysosomes Randomly distributed in Membrane-bound sacs; size varies Contain hydrolytic enzymes If membrane breaks, hydrolytic
cytoplasm for cellular digestive enzymes can destroy cell
system
Microfilaments Near nuclear envelope and Small, solid structure Support cytoskeleton and Consist of actin and myosin
within proximity of approximately 5nm in diameter motility (contractile proteins)
mitotic process
Microtubules Cytoskeleton, near nuclear Hollow cylinder with protofilaments Maintains cell shape, Produced from tubulin
envelope and component surrounding outside tube; motility, and mitotic polymerization; make up
part of centriole near 20-25nm in diameter, variable process mitotic spindles and part of
Golgi body length structure of centriole
Centriole In centrosome near nucleus Cylinder; 150nm in diameter, Serves as insertion point Composed of nine sets of
300-500nm in length for mitotic spindle fibers triplet microtubules

uses the letters CD (cluster designation) and a number a fluid structure of globular proteins floating in lipids. The
following the CD. This common terminology assists in unify- lipids comprise phospholipids and cholesterol arranged in two
ing classification in clinical practice, research efforts, and the layers. The phosphate end of the phospholipid and the
literature (see Chapter 33). Many components found within hydroxyl radical of cholesterol are polar-charged hydrophilic
the cell (e.g., the mitochondria, Golgi apparatus, nucleus, (water-soluble) lipids oriented toward the inner and outer
and endoplasmic reticulum) have similarly constructed mem- surfaces of the cell membrane. The fatty acid portion of the
brane systems. The red blood cell membrane has been widely phospholipid and the steroid nucleus of cholesterol are non
studied and serves as an example of a cell membrane (see polar-charged hydrophobic (water-insoluble) lipids directed
Figure 9-2). toward each other in the center of the cell membrane. Other
To accomplish its many requirements, this cell membrane lipids, such as lipoproteins and lipopolysaccharides, contribute
must be resilient and elastic. It achieves these qualities by being to the membrane structure.
60 PART II Hematopoiesis

Membrane Proteins envelope, which allows communication between the nucleus


Most proteins present in the cell membrane are called glycopro- and the cytoplasm. The number of these pores decreases as the
teins and are found floating in the lipid bilayers.2,3 Two types cell matures.
of proteins, integral and peripheral, have been described in the
cell membrane. Integral proteins may traverse the entirety of the Nucleoli
lipid bilayers and penetrate the outside of the membrane or The nucleus contains one to several nucleoli. The number is
only the cytoplasmic side of the membrane. These transmem- directly proportional to the amount of protein synthesis that
brane proteins are thought to serve as a communication and occurs in the cell. These organelles contain a large amount of
transport system between the cells interior and the external ribosomal RNA, DNA, and other proteins in a loose fibrillar
environment. Peripheral proteins are found only on the inner form. The nucleolus is the site of synthesis of the cells ribo-
cytoplasmic side of the membrane and form the cells cyto somes. Ribosomes consist of two subunits, a large and a small
skeleton. Peripheral proteins also are attached to the cyto one. The subunits are produced in the nucleolus and are trans-
plasmic ends of integral proteins to form a reticular network ported through the nuclear pores for ribosomal assembly and
for maintaining structural integrity and holding the integral protein synthesis.
proteins in a fixed position.

CYTOPLASM
Membrane Carbohydrates
Membrane carbohydrates occur in combination with proteins The cytoplasmic matrix is a homogeneous, continuous,
(glycoproteins) and lipids (glycolipids). The carbohydrate aqueous solution called cytosol. It is the environment in which
portion usually extends beyond the outer cell surface, giving the organelles join and function. These organelles are discussed
the cell a carbohydrate coat often called the glycocalyx. These individually.
carbohydrate moieties function in cell-to-cell recognition and
provide a negative surface charge, surface receptor sites, and Golgi Complex
cell adhesion capabilities.2 The function of the red blood cell The Golgi complex is a system of stacked, membrane-bound,
membrane is discussed in detail in Chapter 9. flattened sacs referred to as cisternae that are involved in modi-
fying, sorting, and packaging macromolecules for secretion or
delivery to other organelles. The number of stacked cisternae
NUCLEUS
ranges from 6 to 30 per cell, depending on the cell type. The
The nucleus is composed of three components: the chromatin, Golgi complex is normally located next to the nucleus. The
the nuclear envelope, and the nucleoli. It is the control center Golgi complex is horseshoe shaped and usually located in close
of the cell and the largest organelle within the cell. The nucleus proximity to the endoplasmic reticulum. The concave aspect
is composed largely of deoxyribonucleic acid (DNA) and is the has numerous enzymes for synthetic activities. The convex side
site of DNA replication and transcription. It is responsible for is the maturing surface and is where the various products are
the chemical reactions within the cell and the cells reproduc- packaged.2
tive process. The nucleus has an affinity for the basic dyes Membrane-bound vesicles are closely associated with the
because of the nucleic acids contained within it. stacks of cisternae. Some of the vesicles are coated and bud off
for transport to other areas of the cell. The Golgi complex
Chromatin directs traffic in the cell. The exact mechanism of macromole-
The chromatin consists of nucleic acids and proteins. The pro- cule modification and sorting is still being defined, although
teins are the histones, which are negatively charged, and the it is clearly a responsibility of the Golgi complex.
nonhistones, which are positively charged. Chromatin has
been divided into two types: (1) the heterochromatin, which is Endoplasmic Reticulum
represented by the more darkly stained, condensed clumping The endoplasmic reticulum (Figure 6-3) is a lacelike network
pattern and is the genetically inactive area of the nucleus, and found throughout the cytoplasm of cells and appears as flat-
(2) the euchromatin, which has diffuse, uncondensed chroma- tened sheets, sacs, and tubes of membrane. The endoplasmic
tin and is the genetically active portion of the nucleus where reticulum subdivides the cytoplasm into various compart-
ribonucleic acid (RNA) transcription occurs. This genetic mate- ments. The outer membrane of the nuclear envelope is in
rial is loosely coiled and turns a pale blue when stained with continuity with the endoplasmic reticulum membrane and
Wright stain. specializes in making and transporting lipid and membrane
proteins.
Nuclear Envelope Rough endoplasmic reticulum has a studded look on its
Surrounding the nucleus is a nuclear envelope consisting of an outer surface caused by the presence of ribosomes engaged in
inner and an outer membrane. The outer membrane is con- the synthesis of proteins. The amount of endoplasmic reticu-
tinuous with an extension of the endoplasmic reticulum. lum found within a cell is proportional to the protein produc-
Between the two membranes is a diaphragm approximately tion required by the cell. More endoplasmic reticulum is
50nm in thickness that is continuous with the lumen of the necessary for increased protein synthesis. Smooth endoplasmic
endoplasmic reticulum. Nuclear pores penetrate the nuclear reticulum does not contain ribosomes and may serve as storage
CHAPTER 6 Cellular Structure and Function 61

Smooth
endoplasmic
reticulum
Rough
endoplasmic
reticulum
Ribosomes

Figure 6-3 Endoplasmic reticulum.

Outer membrane

Oxidation enzymes

Matrix space
Inner membrane

Figure 6-4 Mitochondrion.

sites for the newly synthesized protein. Also, it has been sug- synthesis. This is accomplished with the assistance of transfer
gested as a site for steroid hormone production and synthesis RNA, for amino acid transport to the ribosome, and messenger
of lipid substances. Its function varies with the type of cell. RNA, which provides the necessary information for the
Additional activities of the smooth endoplasmic reticulum sequencing order of the amino acids for each protein.5
include detoxification or inactivation of harmful compounds
or drugs.4 Mitochondria
The existence of mitochondria (Figure 6-4) within the cell has
Ribosomes been known since the nineteenth century, and their function
Ribosomes are small particles composed of nearly equal is now clearly defined. Structurally, the mitochondrion has a
amounts of protein and ribosomal RNA. Ribosomes are found continuous outer membrane. Running parallel to the outer
free in the cytoplasm, on the surface of rough endoplasmic membrane is an inner membrane that invaginates at various
reticulum, and in the nucleus and nucleoli of a cell. These intervals, giving the interior a shelflike or ridgelike appearance.
bodies may exist singly (monoribosome) or form chains (poly- These internal ridges, termed cristae mitochondriales, are where
ribosomes). The more ribosomes present within the cell, the oxidative enzymes are attached. The two membranes differ
more protein production and the more basophilia observed chemically; the inner membrane has a higher protein content
with Wright staining. Ribosomes serve as the site of protein and a lower lipid content. The convolution of the inner
62 PART II Hematopoiesis

membrane increases the surface area to enhance the respiratory Microtubules have several functions. Their contribution to
capability of the cell. The interior of the mitochondrion con- the cytoskeleton helps maintain the cells shape and the move-
sists of a homogeneous material known as the mitochondrial ment of some intracellular organelles. The microtubule makes
matrix, which contains many enzymes for the extraction of up the mitotic spindle fibers and the centrioles during mitosis.11
energy from nutrients. In the peripheral smear, the microtubules and microfilaments
The mitochondria are responsible for the metabolic pro- are not visible, but under special conditions the mitotic spin-
cesses of energy-producing reactions and electron transfer dles may cluster and form Cabot rings.7
oxidative reactions. The oxidative systems described within the
mitochondria are the Krebs cycle, the fatty acid cycle, and the Centrioles
respiratory chain.6 Also present in the mitochondria are pro- Centrioles are paired structures consisting of nine bundles of
teins, phosphorylase, ribosomes, and DNA.7 three microtubules each. They are shaped like cylinders and
The mitochondria are capable of self-replication. This serve as insertion points for the mitotic spindle fibers during
organelle has its own DNA and RNA for the mitochondrial metaphase and anaphase of mitosis. The cylinders are 150nm
division cycle. There may be fewer than 100 or up to several in diameter and 300 to 500nm in length. The long axes are
thousand mitochondria per cell. The number is directly related typically at right angles to one another.
to the amount of energy required by the cell.
Mitochondria do not stain with Wright stain, but instead
HEMATOPOIETIC MICROENVIRONMENT
give a negative image or clear impression against the stained
cytoplasm. When in a hyperactive state, these organelles Hematopoiesis occurs predominantly in the bone marrow after
become so swollen that they appear as short white rods against birth. This is a suitable environment for hematopoietic stem
a blue cytoplasmic background. cells, progenitor cells, and marrow precursors. The marrow
environment must provide for stem cell self-renewal, prolifera-
Lysosomes tion, differentiation, and apoptosis. This protective environ-
Lysosomes contain hydrolytic enzymes bound within a mem- ment is made up of stromal cells, which is a broad term for
brane and are involved in the cells intracellular digestive specialized fibroblasts, reticulum cells, endothelial cells, adipo-
process. The membrane prevents the enzymes from attacking cytes, lymphocytes, and macrophages. Many extracellular
the protein, nucleic acids, mucopolysaccharides, lipids, and molecules are secreted from these cells, including collagen,
glycogen within the cell.8 The enzymes become active when glycoproteins, (fibronectin and thrombospondin), and glycos-
lysosomes bind to the phagocytic vacuole and the membrane aminoglycans. These help form the extracellular matrix that is
ruptures, which allows the escape of the hydrolytic enzymes so critical for the support of cell growth and function in the
into the phagosome. bone marrow microenvironment. Hematopoietic progenitor
With Wright stain, lysosomes are visualized as granules. cells have many receptors for cytokines and adhesion mole-
When stained, many of the granules appear azurophilic, unless cules. One purpose of these receptors is to provide a mecha-
they are too small to be visualized under the light microscope. nism for attachment to extracellular matrix. This provides an
Special staining techniques are required to indicate the pres- avenue for cell-cell interaction, which is essential for regulated
ence of the smaller granules. hematopoiesis. Several adhesion subgroups of receptors help
perform this function. Some examples of the subgroups are
Microfilaments integrins, immunoglobulins, and selectins.12
Microfilaments are solid structures approximately 5nm in Stromal cells also secrete many different growth factors
diameter and consist of actin and myosin proteins. These fibrils required for stem, progenitor, and precursor cell survival.
or groups of fibrils are located near the nuclear envelope or in Growth factors participate in complex processes to regulate the
the proximity of the nucleus and assist in cell division. They proliferation and differentiation of progenitor and precursor
also are present near the membrane for cytoskeletal support cells. Growth factors must bind to specific receptors on their
and motility. Intermediate filaments are the most durable target cells to exert their effect. Most growth factors are pro-
element of the cytoskeleton and provide structural stability for duced by cells in the hematopoietic microenvironment and
the cell, especially during stress.9 Intermediate filaments also exert their effects in local cell-cell interactions. One growth
are detectable and identifiable as cancer markers.4 factor, erythropoietin, has a hormone-type stimulation in that
it is produced in another location (kidney) and exerts its effect
Microtubules on erythroid progenitors in the bone marrow. An important
Microtubules are approximately 25nm in diameter and vary feature of growth factors is their use of synergism to stimulate
in length. These organelles are organized from tubulin through a cell to proliferate or differentiate.12 In other words, several
self-assembly. The tubulin polypeptides form protofilaments. different growth factors work together to generate a more effec-
Usually 13 protofilaments are lined up in parallel rows of tive response. Growth factors are very specific for their corre-
hollow spheres.9,10 This arrangement gives the microtubules sponding receptors on target cells. The receptors are typically
structural strength. A variety of conditions cause microtubules glycoproteins that traverse the membrane. The intracellular
to become disorganized and disappear, especially after mitosis. portion of the receptors associate with protein tyrosine kinase
Tubulin can polymerize and reform the microtubule as needed. and the Janus kinase family, which initiates the cell signaling
CHAPTER 6 Cellular Structure and Function 63

pathways. This will ultimately interact with the DNA in the apoptosis (programmed cell death). A second DNA check takes
nucleus for gene expression and proliferation.13,14 place after DNA synthesis. This checkpoint occurs during G2
and its purpose is to verify that replication took place without
error or damage. If abnormal or malformed replication
CELL CYCLE
occurred, then mitosis is blocked. The last checkpoint is during
The cell cycle is a biochemical and morphologic four-stage mitosis at the time of metaphase. Here the attachment of chro-
process through which a cell passes when it is stimulated to mosomes to the mitotic spindle, the integrity of the spindle
divide (see Figure 31-3). These stages are G1 (gap 1), S (DNA apparatus, and the alignment of the spindles are checked.
synthesis), G2 (gap 2), and M (mitosis). G1 is a period of cell Tumor suppressor genes play a significant role in the proper
growth and synthesis of components necessary for replication. function of the checkpoints. One of the first tumor suppressor
G1 lasts about 10 hours. In the S stage, DNA replication takes genes recognized was p53. The p53 gene detects DNA damage
place, a process requiring about 9 hours. In G2, the nucleus during G1. It can also assist in triggering apoptosis. Multiple
contains tetraploid DNA, and the cell volume seems to help tumor suppressor genes have been described.15,16 When these
trigger the onset of mitosis. DNA is also checked for damage. genes are mutated or deleted, abnormal cells are allowed to go
G2 takes approximately 4 hours. The time spent in each stage through the cell cycle and replicate. Some of these cells simply
can be variable, but mitosis takes approximately 1 hour. During malfunction, but others form neoplasms.17 Inhibitors of the
mitosis cell division occurs. During G0 (quiescence) the cell is cyclin/CDK complexes also play a primary role in cell cycle
not actively in the cell cycle. regulation.13
When the cell receives the signal to proliferate, a number of
biochemical reactions take place. Cell cycle control is under
APOPTOSIS
the direction of cyclin and cyclin-dependent kinases (CDKs).
This cyclin/CDK complex can phosphorylate key substrates Since the concept of programmed cell death or apoptosis was
that assist the cell through cell cycle. Cyclin is named appro- first described many far-reaching purposes of this process have
priately, because the concentration of the cyclin/CDK complex been recognized. Not only does apoptosis limit the number of
moves the cell through the different stages of the cell cycle. G1 cells produced, but its role includes control of organ growth,
begins with a combination of the cyclin D family (D1, D2, D3) which prevents the progression of malignant and abnormal
partnered with CDK4 and CDK6. To transition the cell from cells through the cell cycle. Apoptosis also serves as a defense
G1 to S, cyclin E increases and binds to CDK2, producing the mechanism by removing cells affected by a virus.18
cyclin E/CDK2 complex. In the S stage cyclin E decreases and
cyclin A complexes with CDK2 (cyclin A/CDK2). This combi-
nation takes the cell through the S stage. At the end of S stage, APOPTOSIS
(single cell)
cyclin A starts to activate CDK1 and partners with it. The cyclin
A/CDK1 complex takes the cell through the S stage into G2. For
mitosis to occur, cyclin B must replace cyclin A and bind to
CDK1 in the cyclin B/CDK1 complex. This complex takes the Normal cell
cell through the intricate process of mitosis.13
Mitosis is divided into five phases. Interphase is the first
phase and correlates with G1, S, and G2. Next is prophase, during
which the cell condenses the chromosomes. In prometaphase
the centrosomes move to opposite poles. In metaphase, the
chromosomes are aligned on microtubular spindles. Anaphase
is when the sister chromatids segregate. During telophase, the
Nuclear changes
chromatids move to opposite poles and the cell divides. Nuclear
division is referred to as karyokinesis and cytoplasmic division Cytoplasmic
is cytokinesis. The purpose of the cell cycle is to replicate fragmentation
DNA once during the S stage and distribute identical chromo-
some copies equally to both daughter cells during mitosis.4 Macrophage

Regulation of the Cell Cycle Apoptotic


A regulatory mechanism is needed to prevent abnormal or bodies
mutated cells from going through the cell cycle and producing
an abnormal clone. The cell cycle is a complicated and involved
Figure 6-5 Apoptosis manifests as single cell death. The nucleus con-
process that has numerous stages and biochemical reactions denses and karyorrhexis (fragmentation) occurs. The cell fragments (apop-
that can malfunction. There are several checkpoints or restric- totic bodies) containing parts of the nucleus and functioning cytoplasmic
tion points in the cell cycle at which the level of nutrients and organelles are ultimately engulfed by phagocytic cells, such as macrophages.
DNA is evaluated. The first checkpoint is late in G1. The check- (From Damjanov I: Pathology for the health professions, ed 3, St Louis, 2006,
point makes the cell wait for DNA repair to occur or it initiates Saunders.)
64 PART II Hematopoiesis

There are two major mechanisms for cell death. One is of the death receptor pathway. The third is cell-damaging
apoptosis and the other is necrosis. Apoptosis is internal, stress.
whereas necrosis is caused by external influences. Apoptosis Many proteins have been described that are proapoptotic,
occurs due to the activation of intracellular proteins and trig- and several others have been identified that act as inhibitors to
gering of a regulated cell death. Caspases are the intracellular apoptosis. The BCL-2 family of proteins are antiapoptotic and
proteins that are activated, and this activation occurs through the BAX, BAK, and BAD are examples of proapoptotic proteins.
two different pathways. One pathway involves membrane The ratio of these intracellular proteins plays a primary role in
proteins such as Fas ligand and tumor necrosis factor. This the mechanism for regulating apoptosis. Any dysregulation,
is the death receptor pathway. The second pathway entails mutation, or translocation can cause inhibition or overexpres-
the mitochondrial release of cytochrome c, which in turn binds sion of apoptotic proteins, which can lead to hematopoietic
to Apaf-1. The cytochrome c/Apaf-1 complex is referred to as malignancies or malfunctions.12,17
an apoptosome and results in activation of caspase-9, which The morphologic manifestation of apoptosis is shrinkage of
triggers a cascade leading to apoptosis. This pathway may be the cell. The nucleus and cytoplasm condense. The borders of
the one involved in cellular response to chemotherapy or the cell begin to bud off. Macrophages eliminate the cell
irradiation.12 through phagocytosis (Figure 6-5). No inflammatory response
The initiation of apoptosis typically occurs from three occurs.
possible sources. The first is an abnormal hematopoietic The morphologic manifestation of necrosis is a swelling of
microenvironment in the bone marrow and/or decreased levels the cell with damage of the plasma membrane and lysis of the
of growth factors (see Chapter 7). The second is stimulation cell. An inflammatory response then occurs.17

SUMMARY
Cells are building blocks of the living organism and provide the Lysosomes contain hydrolytic enzymes involved in the cells intra-
basis for all life processes. cellular digestive process.
The nucleus serves as a control center: it directs, transmits infor- The cell cycle involves four active stages: G1 (gap 1), S (DNA
mation, and maintains the cell. synthesis), G2 (gap 2), and M (mitosis).
The nucleolus is the site of synthesis and processing of ribosomal The cell cycle is under the direction of cyclin and CDKs.
RNA. Checkpoints in the cell cycle recognize abnormalities and initiate
The Golgi complex modifies and packages macromolecules for apoptosis.
secretion.
Mitochondria make adenosine triphosphate to supply energy for
the cell.

R E V I E W Q UESTIONS
1. The organelle involved in packaging and trafficking of cel- 4. The nucleus is composed largely of:
lular products is the: a. RNA
a. Nucleus b. DNA
b. Golgi complex c. Ribosomes
c. Mitochondria d. Glycoproteins
d. Rough endoplasmic reticulum
5. Protein synthesis occurs in the:
2. The most common type of protein found in the cell mem- a. Nucleus
brane is: b. Mitochondria
a. Lipoprotein c. Ribosomes
b. Mucoprotein d. Golgi complex
c. Glycoprotein
d. Nucleoprotein 6. The shape of a cell is maintained by which of the
following?
3. The control center of the cell is the: a. Microtubules
a. Nucleus b. Spindle fibers
b. Cytoplasm c. Ribosomes
c. Membrane d. Centrioles
d. Microtubular system
CHAPTER 6 Cellular Structure and Function 65

7. Functions of the cell membrane include all of the follow- 11. The cell cycle is regulated by:
ing except: a. Cyclins and CDKs
a. Interchange of substances b. Protooncogenes
b. Cell-to-cell recognition c. Apoptosis
c. Cellular identification through receptors d. Growth factors
d. Lipid production
12. The transition from the G1 to S stage of the cell cycle is
8. The energy source for cells is the: regulated by:
a. Golgi complex a. Cyclin B/CDK1 complex
b. Endoplasmic reticulum b. Cyclin A/CDK2 complex
c. Nucleolus c. Cyclin D1
d. Mitochondrion d. Cyclin E/CDK2 complex

9. Ribosomes are synthesized by the: 13. Apoptosis is morphologically identified by:


a. Endoplasmic reticulum a. Cellular swelling
b. Mitochondrion b. Nuclear condensation
c. Nucleolus c. Rupture of the cytoplasm
d. Golgi apparatus d. Rupture of the nucleus

10. Euchromatin functions as the:


a. Site of microtubule production
b. Genetically active DNA
c. Support structure for nucleoli
d. Attachment site for centrioles

REFERENCES
1. Wintrobe MM: Blood, pure and eloquent, New York, 1980, 11. Stephens RE, Edds KT: Microtubules: structure, chemistry and
McGraw-Hill. function. Physiol Rev 56:709-777, 1976.
2. Guyton AC, Hall JE: Textbook of medical physiology, ed 11, 12. Hoffbrand JV, Pettit JE, Moss PAH: Essential haematology,
Philadelphia, 2006, Saunders. ed 4, Oxford, 2004, Blackwell.
3. Steinberg MH, Benz EJ, Adewoye AH, et al: Pathobiology of 13. Williams JL: Cellular homeostasis. In McKenzie SB, Williams
the human erythrocyte and its hemoglobins. In Hoffman R, JL, editors: Clinical laboratory hematology, ed 2, New York,
Furie B, Benz EJ, et al, editors: Hematology: basic principles and 2010, Pearson, pp 15-18.
practice, ed 5, Philadelphia, 2009, Churchill Livingstone. 14. Kaushansky K: Hematopoietic stem cells, progenitors, and
4. Becker WM, Kleinsmith IJ, Hardin J: The world of the cell, cytokines. In Lichtman MA, Beutler E, Kipps TJ, et al, editors:
ed 7, Menlo Park, Calif, 2009, Benjamin Cummings. Williams hematology, ed 7, New York, 2006, McGraw-Hill,
5. Koss LG, editor: Koss diagnostic cytology and its histopathologic pp 201-220.
bases, vol 1, ed 5, Philadelphia, 2005, Lippincott Williams & 15. Important tumor suppressors. Emory University CancerQuest
Wilkins. website. Available at: http://www.cancerquest.org/
6. Prebble JN: Mitochondria, chloroplasts and bacterial membranes, index.cfm?page=52&lang=english. Accessed March 9, 2010.
New York, 1981, Longman. 16. Additional tumor suppressors. Emory University Cancer-
7. Bessis M: Blood smears reinterpreted, Berlin, 1977, Springer Quest website. Available at: http://www.cancerquest.org/
International. index.cfm?page=781. Accessed March 9, 2010.
8. De Duve C, Wattiaux R: Functions of lysosomes. Annu Rev 17. Mendelsohn J, Howley PM, Israel MA, et al: The molecular
Physiol 28:435-492, 1966. basis of cancer, ed 3, Philadelphia, 2008, Saunders.
9. Alberts B, Johnson A, Lewis J, et al: Molecular biology of the 18. Teodoro JG, Branton PE: Regulation of apoptosis by viral
cell, ed 5, New York, 2007, Garland. gene products. J Virol 71(3):1739-1746, 1997.
10. Alberts B, Bray D, Hopkin K, et al: Essential cell biology, ed 3,
New York, 2009, Garland.

ADDITIONAL RESOURCE
Lodish H, Berk A, Kaiser C, et al: Molecular cell biology, ed 6,
New York, 2008, WH Freeman.
7 Hematopoiesis
Larry Smith

OUTLINE OBJECTIVES
Hematopoietic After completion of this chapter, the reader will be able to:
Development
Mesoblastic Phase 1. Define hematopoiesis. hematopoietic progenitor cells, including nonspecific
(Yolk Sac Phase) 2. Discuss the evolution and formation of blood cells and lineage-specific factors.
Hepatic Phase from embryo to fetus to adult, including anatomic 7. Describe general morphologic changes that occur
Medullary (Myeloid) Phase sites and cells produced. during cell maturation.
Adult Hematopoietic 3. Predict the likelihood of encountering active marrow 8. Define apoptosis and discuss the relationship
Tissue from biopsy sites when given the patients age. between apoptosis, growth factors, and stem cell
Bone Marrow 4. Relate normal and abnormal hematopoiesis to the differentiation.
Red Marrow various organs involved in the hematopoietic process. 9. Discuss the roles of various cytokines and hema
Marrow Circulation 5. Explain the stem cell theory of hematopoiesis, topoietic growth factors in the process of
Hematopoietic
including the names of various stem cells and pro- hematopoiesis.
Microenvironment
genitor cells and their lineage associations. 10. Discuss therapeutic applications of cytokines and
Liver
Spleen 6. Discuss the roles of hematopoietic growth factors hematopoietic growth factors.
Lymph Nodes in differentiation and maturation sequences of
Thymus
Stem Cells and Cytokines
Stem Cell Theory
Stem Cell Cycle Kinetics
HEMATOPOIETIC DEVELOPMENT
Stem Cell Phenotypic and Hematopoiesis is a continuous, regulated process of blood cell production that includes cell renewal,
Functional proliferation, differentiation, and maturation. These processes result in the formation, development,
Characterization and specialization of all of the functional blood cells that are released from the bone marrow to the
Cytokines and Growth
circulation. In adults, all of these processes are restricted primarily to the bone marrow. During fetal
Factors
development, hematopoiesis occurs in different areas of the developing fetus. This process has been
Colony-Stimulating
Factors divided into three phases: the mesoblastic phase, the hepatic phase, and the medullary phase.
Stem Cell Factor, Flt3
Ligand, Granulocyte/ Mesoblastic Phase (Yolk Sac Phase)
Monocyte Colony- Hematopoiesis is considered to begin around the nineteenth day of embryologic development after
Stimulating Factor, fertilization. Progenitor cells of mesenchymal origin migrate from the aorta-gonad-mesonephros
Interleukin-3, and region of the developing aorta-splanchnopleure to the yolk sac.1 The cells arising from the aorta-gonad-
Interleukin-6 mesonephros region give rise to hematopoietic stem cells (HSCs), but not to primitive erythroblasts.
Interleukins The primitive erythroblasts found in the yolk sac arise from mesodermal cells, which initially line the
Lineage-Specific cavity of the yolk sac. These primitive cells migrate from the periphery into the central cavity of the
Hematopoiesis
yolk sac, where they develop into primitive erythroblasts.2-5 The remaining cells surrounding the cavity
Erythropoiesis
of the yolk sac are called angioblasts and form the future blood vessels.2-5 The yolk sac phase of hema-
Leukopoiesis
Megakaryopoiesis topoiesis is characterized by the development of primitive erythroblasts that produce measurable
Analytical and amounts of hemoglobins, including Portland, Gower-1, and Gower-2 (see Chapter 10). Yolk sac
Therapeutic hematopoiesis does not contribute significantly to definitive hematopoiesis.4 This phase of hemato-
Applications poiesis occurs intravascularly, or within a developing blood vessel.

Hepatic Phase
The hepatic phase of hematopoiesis begins at 4 to 5 gestational weeks and is characterized by recog-
nizable clusters of developing erythroblasts, granulocytes, and monocytes.5 The developing erythro-
blasts signal the beginning of definitive hematopoiesis with a decline in primitive hematopoiesis of

66
CHAPTER 7 Hematopoiesis 67

Cellularity (%)

100

Bone marrow
80
Vertebra
Yolk sac Liver
60
Sternum
1 2 3
40 Femur

Tibia Rib
20
Spleen Lymph nodes

0
1 2 3 4 5 6 7 8 9 10 20 30 40 50 60
Fetal months Birth Age in years
Sites of hematopoiesis
1 Mesoblastic
2 Hepatic
3 Myeloid

Figure 7-1 Sites of hematopoiesis.

the yolk sac. In addition, lymphoid cells begin to appear.6,7 colony-stimulating factor (G-CSF), granulocyte-monocyte
Hematopoiesis during this phase occurs extravascularly, with colony-stimulating factor (GM-CSF), fetal hemoglobin, Hb A2,
the liver remaining the major site of hematopoiesis during fetal and adult hemoglobin can be detected. In addition, cells at
life and retaining activity until 1 to 2 weeks after birth. Hema- various stages of maturation can be seen in all three lineages.
topoiesis in the aorta-gonad-mesonephros region and the yolk
sac disappears during this stage. Hematopoiesis in the fetal
ADULT HEMATOPOIETIC TISSUE
liver reaches its peak by the third month of development
(Figure 7-1). The developing spleen, kidney, thymus, and In adults, hematopoietic tissue is involved in the proliferation
lymph nodes contribute to the hematopoietic process during and maturation of blood cells. Numerous organs and tissues
this phase. The thymus, the first fully developed organ in the contribute to this process, including the bone marrow, lymph
fetus, becomes the major site of T-cell production, whereas the nodes, spleen, liver, and thymus. The bone marrow contains
kidney and spleen produce B cells. developing erythroid, myeloid, megakaryocytic, and lymphoid
Production of megakaryocytes also begins during the cells. The tissues where lymphoid development occurs are
hepatic phase. The spleen gradually decreases granulocytic pro- divided into primary and secondary lymphoid tissue. Primary
duction and involves itself solely in lymphopoiesis. During the lymphoid tissue consists of the bone marrow and thymus and
hepatic phase, detectable levels of hemoglobin (Hb) F, Hb A, is where T and B cells are derived. Secondary lymphoid tissue,
and Hb A2 may be present.8 where lymphoid cells become competent, consists of the
spleen and lymph nodes and gut-associated lymphoid tissue.
Medullary (Myeloid) Phase
During the fifth month of fetal development, hematopoiesis Bone Marrow
begins in the developing bone marrow cavity. This transition Bone marrow, one of the largest organs in the body, is defined
is called medullary hematopoiesis because it occurs in the medulla as the tissue located within the cavities of the cortical bones.
or inner part of the bone marrow. During this phase, mesen- These cavities consist of trabecular bone resembling a honey-
chymal cells, which are a type of embryonic tissue, migrate into comb. Normal bone marrow located within these cavities
the core of the bone and differentiate into skeletal and hema- consists of two types of marrow: (1) red marrow, which is hema-
topoietic blood cells.9,10 Hematopoietic activity, especially topoietically active marrow, and (2) yellow marrow, represent-
myeloid activity, is apparent during this stage of development, ing hematopoietically inactive marrow composed primarily of
and the myeloid-to-erythroid ratio approaches the adult level adipocytes (fat cells). Red marrow in adults is found in the
of 3:1 by 21 weeks of gestation.11 By the end of the sixth month, sternum, skull, scapulae, vertebrae, ribs, pelvic bones, and
the bone marrow becomes the primary site of hematopoiesis. proximal ends of the long bones. Normal adult bone marrow
Measurable levels of erythropoietin (EPO), granulocyte has approximately equal amounts of red and yellow marrow.
68 PART II Hematopoiesis

A central space is created within the cavities of these bones marrow in cases of increased demand on the bone marrow.8
by resorption of cartilage and endosteal bone. Mesenchymal Such cases might be excessive blood loss or increased erythro-
cells migrate into the space and eventually differentiate into cyte destruction in the bone marrow by toxic chemicals or
three cell types, which give rise to the blood and bone marrow irradiation.
matrix cells (reticular cells and adipose tissue). Reticular cells
are formed on the exterior surfaces of the venous sinuses and Red Marrow
extend long, narrow branches into the perivascular space, creat- The red marrow is composed of extravascular cords that contain
ing a meshlike network; this provides a supportive skeletal all of the developing blood cell lineages, stem and progenitor
network for developing hematopoietic cells, macrophages, and cells, adventitial cells, and macrophages (Figures 7-3 and 7-4).
mast cells.10 During infancy and early childhood, the bone The cords are separated from the lumen of the sinusoids by
marrow consists primarily of red active marrow. Between 5 and endothelial and adventitial cells and are located between the
7 years of age, adipocytes become more abundant and begin trabeculae of spongy bone. Trabeculae are projections of calci-
to occupy the spaces in the long bones previously dominated fied bone radiating out from the cortical bone into the marrow
by active marrow. The process of replacing the active marrow space and provide support for the developing marrow. The
by adipose tissue (yellow marrow) during development is hematopoietic cells tend to develop in specific niches within
called retrogression and eventually results in restriction of the the cords. Normoblasts develop in small clusters adjacent to the
active marrow to the flat bones, sternum, vertebrae, pelvis, ribs, outer surfaces of the vascular sinuses (Figure 7-5); in addition,
skull, and proximal portion of the long bones (Figure 7-2). some normoblasts are found surrounding iron-laden macro-
Areas located within the bone marrow cavity where red marrow phages (Figure 7-6). Megakaryocytes are located close to the
has been replaced by yellow marrow consist of an mixture of vascular walls of the sinuses, which facilitates the release of
adipocytes, undifferentiated mesenchymal cells, and macro-
phages. Inactive yellow marrow is also scattered throughout
active red marrow and is capable of reverting back to active Hematopoietic
cords
Central
sinus
Skull
Central
artery

Bone
Proximal end of Endosteum
large bones
Sternum
Vertebrae

Axial skeleton
Iliac crest Megakaryocyte

Erythrocytic Granulocytes
cells

Adventitial
cells

Fat Endothelial
cell cells

Figure 7-2 The adult skeleton, in which darkened areas depict active red Figure 7-3 Graphic illustration of the arrangement of the extravascular
marrow hematopoiesis. area in hematopoietic tissue.
CHAPTER 7 Hematopoiesis 69

Hematopoietic tissue
Adventitial
cells

Bone

Megakaryocyte

Adventitial
Adipocytes cells Arteriole

Adipocytes
Erythroid
Processes precursors
of adventitial
cells

Bone

Figure 7-4 Fixed and stained bone marrow biopsy specimen (hematoxylin Figure 7-5 Fixed and stained bone marrow biopsy specimen (hematoxylin
and eosin stain, 100). The extravascular tissue consists of blood cell precur- and eosin stain, 400). Hematopoietic tissue reveals areas of granulopoiesis
sors and various tissue cells with scattered fat tissue. A normal adult bone (lighter-staining cells) and erythropoiesis (darker-staining nuclei). One
marrow displays 50% tissue and 50% fat. megakaryocyte can be seen. Adventitial cells and their processes give
support to the hematopoietic cells; they also guard apertures of the basement
membrane.
platelets into the lumen of the sinusoids. Immature myeloid
(granulocytic) cells through the metamyelocyte stage are located
deep within the cords. As these maturing granulocytes proceed through pores in the endothelial cytoplasm and are released
along their differentiation pathway, they move closer to the into the circulation.12
vascular sinuses.9
The mature blood cells of the bone marrow eventually enter Marrow Circulation
the peripheral circulation by a process that is not well under- The nutrient and gas requirements of the marrow are supplied
stood. Through a highly complex interaction between the by the nutrient and periosteal arteries, which enter via the bone
maturing blood cells and the sinus wall, blood cells pass foramina. The nutrient artery supplies blood only to the
between layers of adventitial cells that form a discontinuous marrow.10 It coils around the central longitudinal vein, which
layer along the abluminal side of the bone marrow sinus. passes along the bone canal. In the marrow cavity, the nutrient
Adjacent to the layer of adventitial cells is a basement membrane artery divides into ascending and descending branches that
followed by a continuous layer of endothelial cells on the luminal also coil around the central longitudinal vein. The arteriole
side of the bone marrow sinus. These adventitial cells are branches that enter the inner lining of the cortical bone (endo
described as reticular cells and extend long cytoplasmic processes steum) form sinusoids (endosteal beds), which connect to perio
into the marrow cords. The extensions of these reticular fila- steal capillaries that extend from the periosteal artery.13 The
ments form a meshwork that provides support for the develop- periosteal arteries provide nutrients for the osseous bone and the
ing hematopoietic cells. The adventitial cells are capable of marrow. Their capillaries connect to the venous sinuses located
contracting, which allows mature blood cells to pass through in the endosteal bed, which empty into a larger collecting sinus
the basement membrane and interact with the endothelial layer. that opens into the central longitudinal vein.14 Blood exits the
As blood cells come in contact with endothelial cells, they marrow via the central longitudinal vein, which runs the length
bind to the surface via a receptor-mediated process. Cells pass of the marrow. The central longitudinal vein exits the marrow
70 PART II Hematopoiesis

are located throughout the marrow space.17 Other cells involved


in cytokine production include endothelial cells, adipocytes,
and fibroblasts. Osteoblasts are bone-forming cells, and osteo
clasts are bone-resorbing cells. Reticular cells are associated with
the formation of reticular fibers that form a lattice that supports
the vascular sinuses and developing hematopoietic cells.
Stromal cells are believed to be derived from fibroblasts. They
play a role in support and regulation of hematopoietic stem/
progenitor cell survival and differentiation.15
The extracellular matrix of the bone marrow contains
proteoglycans or glycosaminoglycans, fibronectin, collagen, laminin,
hemonectin, and thrombospondin.15 Proteoglycans are expressed
on the endothelial cell surface and mediate progenitor binding
to the stroma. Fibronectin, collagen, laminin, hemonectin, and
thrombospondin function as adhesion molecules, promoting
the adhesion of HSCs to the extracellular matrix.

Liver
The liver plays a significant role in hematopoiesis beginning
around the second trimester and serves as the major site of
blood cell production during the hepatic stage of hemato
poiesis. In adults, the liver has many cellular production
functions, including synthesizing various transport proteins,
storing essential minerals and vitamins that are used in the
synthesis of deoxyribonucleic acid (DNA) and ribonucleic acid
(RNA), conjugating bilirubin from hemoglobin degradation,
and transporting bilirubin to the small intestine for eventual
excretion.
The liver consists of two lobes situated beneath the dia-
phragm in the abdominal cavity. The position of the liver with
Figure 7-6 Bone marrow aspirate smear (Wright-Giemsa stain, 500). regard to the circulatory system is optimal for gathering, trans-
Macrophage with extensive iron-laden cytoplasm, surrounded by developing
ferring, and eliminating substances via the bile.18 Anatomically,
erythroid precursors.
liver cells are arranged in radiating hepatic lobules emanating
from a central vein (Figure 7-7). Adjacent to the longitudinal
through the same foramen where the nutrient artery enters. lobes of the liver and separated only by a small space are sinu-
Hematopoietic cells located in the endosteal bed receive their soids, which are lined by two types of cells: Kupffer cells and
nutrients from the nutrient artery. epithelial cells. Kupffer cells are macrophages, removing cel-
lular and foreign debris from the blood that circulates through
Hematopoietic Microenvironment the liver; they also are responsible for protein synthesis.19 The
The hematopoietic inductive microenvironment plays an important epithelial cells are arranged in the lining so as to be separated
role in stem cell differentiation and proliferation.15,16 It is from one another by a noncellular area; this arrangement
responsible for supplying a semifluid matrix, which serves as allows plasma to have direct access to the hepatocytes. This
an anchor for the developing hematopoietic cells. The matrix unusual organization of the liver and its location in the body
is responsible for maintaining differentiation and proliferation enables it to be involved in many varied functions.
and provides a supporting tissue in the hematopoietic induc-
tive microenvironment. Stromal cells in the matrix are of several Liver Pathophysiology
types: (1) endothelial cells, (2) adipocytes, (3) macrophages, The liver is often involved in blood-related diseases. In por
(4) osteoblasts, (5) osteoclasts, and (6) reticular cells (fibro- phyrias, the liver exhibits enzymatic deficiencies that result in
blasts). Endothelial cells are broad flat cells that form a single the accumulation of the various intermediary porphyrins.
continuous layer along the inner surface of the bone marrow In severe hemolytic anemias and red blood cell (RBC) dyspla-
sinus.15 They regulate the flow of particles entering and leaving sias, the conjugation of bilirubin and the storage of iron are
hematopoietic spaces. Adipocytes are large cells with a single fat increased. The liver sequesters membrane-damaged RBCs and
vacuole; they secrete various steroids that influence erythro removes them from the circulation. The liver is capable of
poiesis and maintain bone integrity.14 They also play a role in extramedullary hematopoietic production in case of bone
regulating the volume of the marrow in which active hemato- marrow shutdown.14 It is directly affected by storage diseases
poiesis occurs.14 Macrophages function in phagocytosis and of the monocyte/macrophage (Kupffer) cells as a result of enzy-
secretion of various cytokines that regulate hematopoiesis and matic deficiencies that cause hepatomegaly with ultimate
CHAPTER 7 Hematopoiesis 71

Fat- Sinusoid
storing Central Liver endothelial
cells Sinusoid vein plates cell

Distributing Hepatic Portal Bile Kupffer


vein artery vein duct cells

Inlet Bile
venule canaliculi

Figure 7-7 Three-dimensional schematic of the normal liver.

dysfunction of the liver (Gaucher disease, Niemann-Pick macrophages, and dendritic cells. Aggregates of lymphocytes
disease, Tay-Sachs disease; see Chapter 28). surround splenic arteries that pass through these germinal
centers. Adjacent to the splenic arteries is a region called the
Spleen periarteriolar lymphatic sheath. This area consists of lymphoid
The spleen is the largest lymphoid organ in the body. The nodules containing primarily B lymphocytes. Activated B lym-
spleen is located directly beneath the diaphragm behind phocytes are found in the germinal centers.19
the fundus of the stomach in the upper left quadrant of the The marginal zone surrounds the white pulp and forms a
abdomen. It is vital but not essential for life and functions as reticular meshwork containing blood vessels, macrophages,
an indiscriminate filter of the circulating blood. In a healthy and specialized B cells. The red pulp is composed primarily of
individual, the spleen contains about 350mL of blood.18 vascular sinusoids and sinuses separated by cords of tissue
The exterior surface of the spleen is surrounded by a layer (cords of Billroth) containing specialized macrophages that are
of peritoneum and inwardly by a connective tissue capsule. loosely connected to the dendritic process, creating a sponge-
The capsule projects inwardly, forming trabeculae that divide like region that functions as a filter for blood passing through
the spleen into discrete regions. Located within these regions the region.19 As RBCs pass through the cords of Billroth, there
are three types of splenic tissue: (1) white pulp, (2) red pulp, is a decrease in the flow of blood, which leads to stagnation
and (3) a marginal zone. The white pulp consists of scattered and depletion of the RBCs glucose supply. These cells are
follicles with germinal centers containing lymphocytes, subject to increased damage and stress that may lead to their
72 PART II Hematopoiesis

Lymphatic nodule
(containing germinal center) Splenic cords (of red pulp)
White pulp
Periarterial
lymphatic sheath

Trabecular
Venous vein
sinus

Marginal zone

Capsule

Pulp
vein
Venous sinus
in red pulp

Venous sinus
(contiguous to
white pulp)
Lymphatic Periarterial Lymphatic
vessel Central lymphatic nodule
artery sheath
Figure 7-8 Schematic of the normal spleen. (From Weiss L, Tavossoli M: Anatomical hazards to the passage of erythrocytes through the spleen,
Semin Hematol 7:372-380, 1970.)

removal from the spleen. The spleen uses two methods for which the RBCs pass circuitously through the macrophage-
removing senescent RBCs from the circulation: (1) culling, in lined cords before reaching the sinuses. Plasma freely reaches
which the cells are phagocytosed with subsequent degradation the sinuses, but the RBCs have a more difficult time passing
of cell organelles, and (2) pitting, in which splenic macro- through the tiny openings created by the interendothelial junc
phages remove inclusions or damaged surface membrane from tions of adjacent endothelial cells. The combination of the
the circulating RBCs. The spleen synthesizes immunoglobulin slow passage and the continued RBC metabolism creates an
M in the germinal centers, and it serves as a storage site for environment that is acidic, hypoglycemic, and hypoxic. The
platelets. In a healthy individual, approximately 30% of the increased environmental stress on the RBCs circulating through
total platelet count is sequestered in the spleen.20 the spleen leads to possible hemolysis.
The spleen has a rich blood supply, receiving approximately In the rapid-transit pathway, blood cells enter the splenic
350mL/min. Blood enters the spleen through the central artery and pass directly to the sinuses in the red pulp and
splenic artery located at the hilum and branches outward through continue to the venous system to exit the spleen. When spleno
the trabeculae. The branches enter all three regions of the megaly occurs, the spleen becomes enlarged and is palpable.
spleen: the white pulp with its dense accumulation of lympho- This occurs as a result of many conditions, such as chronic
cytes, the marginal zone, and the red pulp. The venous sinuses, leukemias, genetic defects in RBCs, hemoglobinopathies,
which are located in the red pulp, unite and leave the spleen Hodgkin disease, thalassemia, malaria, and the myeloprolifera-
as splenic veins (Figures 7-8 and 7-9).21 tive disorders. Often splenectomy is beneficial in cases of exces-
sive destruction of RBCs, such as severe hereditary spherocytosis,
Spleen Pathophysiology storage disorders, and autoimmune hemolytic anemias, when
As blood enters the spleen, it may follow one of two routes. treatment with corticosteroids does not effectively suppress
The first is a slow-transit pathway through the red pulp in hemolysis.22,23 Splenectomy also may be indicated in severe
CHAPTER 7 Hematopoiesis 73

Figure 7-9 Scanning electron micrograph of the spleen shows erythrocytes (numbered 1 to 6) squeezing through the fenestrated wall in transit
from the splenic cord to the sinus. The view shows the endothelial lining of the sinus wall, to which platelets (P) adhere, along with hairy white cells,
probably macrophages. The arrow shows a protrusion on a red blood cell (5000). (From Weiss L: A scanning electron microscopic study of the spleen,
Blood 43:665, 1974.)

cases of agnogenic myeloid metaplasia associated with spleno- Lymph is filtered by the lymph nodes and exits via the efferent
megaly, severe refractory hemolytic anemia, thrombocytope- lymphatic vessels located in the hilus of the lymph node.25
nia, or qualitative platelet function defect syndromes.22,23 After Similar in structure to the spleen, lymph nodes consist of
splenectomy, platelet and leukocyte counts increase tran- an outer capsule that forms trabeculae and provides support
siently.22 In sickle cell anemia, repeated splenic infarcts caused for macrophages and the predominant population of lympho-
by sickled RBCs trapped in the small-vessel circulation of the cytes. Lymph nodes can be divided into two basic regions: an
spleen cause tissue damage and necrosis, which often results outer region called the cortex and an inner region called the
in autosplenectomy (see Chapter 26). medulla. Trabeculae radiate through the cortex and the medulla,
Hypersplenism is an enlargement of the spleen resulting in dividing the interior of the lymph node into specific areas
some degree of pancytopenia despite the presence of a hyperac- (Figure 7-10). In the cortical region, these areas are known as
tive bone marrow. The most common cause is congestive sple- cortical nodules and contain follicles. These follicles contain foci
nomegaly secondary to cirrhosis of the liver and portal of B-cell proliferation termed germinal centers.9,18 The cortical
hypertension. Other causes include thrombosis, vascular ste- nodules are arranged in circles along the outer cortex region of
nosis, other vascular deformities such as aneurysm of the the lymph node. Located between the cortex and the medulla
splenic artery, and cysts.24 is a region called the paracortex, which contains predominantly
T cells and numerous macrophages. The medullary cords lie
Lymph Nodes toward the interior of the lymph node. These cords consist
Lymph nodes are organs of the lymphatic system located along primarily of B lymphocytes and plasma cells.23 Lymph nodes
the lymphatic capillaries that parallel, but are not part of, the have three main functions: (1) they play a role in the formation
circulatory system. The nodes are bean-shaped structures (1 to of new lymphocytes from the germinal centers, (2) they are
5mm in diameter) that occur in groups or chains at various involved in the processing of specific immunoglobulins, and
intervals along lymphatic vessels. They may be superficial (3) they filter particulate matter, debris, and bacteria entering
(inguinal, axillary, cervical, supratrochlear) or deep (mesen- the lymph node via the lymph.
teric, retroperitoneal). Lymph is the fluid portion of blood that
escapes into the connective tissue and is characterized by a low Lymph Node Pathophysiology
protein concentration and the absence of RBCs. Afferent lym- Lymph nodes, by their nature, are vulnerable to the same
phatic vessels carry circulating lymph to the lymph nodes. organisms that circulate through the tissue. Sometimes
74 PART II Hematopoiesis

Afferent
lymphatic
vessel
Trabecula

Secondary
follicle
Follicle
(primary)

Medullary
sinus

Paracortical
area
(T cells)

Capsule

Efferent Medullary
lymphatic cord
vessel (plasma
cells)

Figure 7-10 Histologic structure of a normal lymph node. The cortical layer is composed mainly of lymphatic nodules, whose germinal centers can be
clearly seen.

increased numbers of microorganisms enter the nodes, over- migrated from the bone marrow. These cells have no identifi-
whelming the macrophages and causing adenitis (infection able surface markers when they enter the thymus but give rise
of the lymph node). More serious is the frequent entry into to T cells that later express surface antigens and move toward
the lymph nodes of malignant cells that have broken loose the medulla. Eventually, they leave the thymus to populate
from malignant tumors. These malignant cells may establish specific regions of other lymphoid tissue, such as the T cell
new growths, which metastasize to other lymph nodes in the dependent areas of the spleen, lymph nodes, and other lym-
same group. phoid tissues that are deficient in immunocompetent T lymphocytes.
It is theorized that the cytoplasmic processes of the epithelial
Thymus reticular cells contain secretory products, thymic hormones,
To understand the role of the thymus in adults, certain thymic factor, and thymic humoral hormones (proteins/
formative intrauterine processes that affect function must peptides extracted from the thymus) that promote differentia-
be considered. First, the thymus tissue originates from endo- tion of pre-T (nonmarked) from mature T lymphocytes. The
dermal and mesenchymal tissue. Second, the thymus is cells that are not marked die in the cortex as a result of apop-
populated initially by lymphocytes from the yolk sac and tosis and are phagocytosed by macrophages before release. The
the liver. This increased population of lymphoid cells physi- medulla contains only 5% mature T lymphocytes and seems
cally pushes the epithelial cells of the thymus apart; however, to be a holding zone for conditioned cells until the cells are
their long processes remain attached to each other by needed by the peripheral lymphoid tissues.26 The thymus also
desmosomes. contains myriad cell types, including B cells, dendritic cells,
At birth, the thymus is an efficient, well-developed organ. eosinophils, neutrophils, and myeloid cells.19
It consists of two lobules, each measuring 0.5 to 2cm in diam- Gross examination indicates that the size of the thymus is
eter. It is located in the upper part of the anterior mediastinum related to age. The thymus weighs 12 to 15g at birth, increases
at about the level of the great vessels of the heart.9,18 It resem- to 30 to 40g at puberty, and decreases to 10 to 15g at later
bles other lymphoid tissue in that the lobules are subdivided ages. It is hardly recognizable in old age, having atrophied
into two areas: the cortex (a peripheral zone) and the medulla (Figure 7-12). The thymus retains the ability to produce new T
(a central zone) (Figure 7-11). Both areas are populated with cells, however, as has been shown after irradiation treatment
the same cellular componentslymphocytes, mesenchymal that may accompany bone marrow transplantation.
cells, reticular cells, and many macrophagesalthough in dif-
ferent proportions. The cortex is characterized by a blood Thymus Pathophysiology
supply system that is unique in that it consists only of capil- Nondevelopment of the thymus during gestation results in the
laries. Its function seems to be that of a waiting zone, which lack of formation of T lymphocytes. Related manifestations
is densely populated with progenitor lymphoid cells that seen in patients with this condition are failure to thrive,
CHAPTER 7 Hematopoiesis 75

Cortex Medulla Epithelial cells and Macrophage Hassall's


dendritic cells corpuscle
Figure 7-11 Schematic diagram of the edge of a lobule of the thymus, showing cells of the cortex and medulla. (From Abbas AK, Lichtman AH, Pober JS:
Cellular and molecular immunology, Philadelphia, 1991, Saunders.)

Trachea

Thyroid
Thyroid

Trachea

Thymus
Thymus
Lungs

Heart Lungs

Heart

B
Figure 7-12 Differences in the size of the thymus of the infant (A) and the adult (B).
76 PART II Hematopoiesis

uncontrollable infections, and death in infancy. Adults TABLE 7-1 Culture-Derived Colony-Forming Units (CFUs)
with thymic disturbance are not affected because they
Abbreviation Cell Line
have developed and maintained a pool of T lymphocytes
for life. CFU-GEMM Granulocyte, erythrocyte, megakaryocyte, monocyte
CFU-E Erythrocyte
CFU-Meg Megakaryocyte
CFU-M Monocyte
STEM CELLS AND CYTOKINES
CFU-GM Granulocyte, monocyte
Stem Cell Theory CFU-BASO Myeloid to basophil
Hematopoiesis is a dynamic and complex developmental CFU-EO Myeloid to eosinophil
process of blood cell production. Blood cell production that CFU-G Myeloid to neutrophil
occurs during the mesoblastic stage of development is referred CFU-pre-T T lymphocyte
to as primitive hematopoiesis, whereas definitive hematopoiesis CFU-pre-B B lymphocyte
begins during the fetal liver stage and continues through
adult life.
In 1961 Till and McCulloch27 conducted a series of experi-
ments in which they irradiated spleens and bone marrows of normal bone marrow are precursor cells at various stages of
mice, creating a state of aplasia. These aplastic mice were given maturation.
an intravenous injection of marrow cells. Colonies of HSCs HSCs are directed to one of three possible fates: (1) self-
were seen 7 to 8 days later in the spleens of the irradiated renewal, (2) differentiation, or (3) apoptosis.29 When the HSC
(recipient) mice. These colonies were called colony-forming divides, it gives rise to two identical daughter cells. Both daugh-
unitsspleen (CFU-S). These investigators later showed that ter cells may follow the path of differentiation, leaving the
these colonies were capable of self-renewal and the production stem cell pool (symmetric division), or one daughter cell may
of differentiated progeny. The CFU-S represents what we now return to the stem cell pool and the other daughter cell may
refer to as committed myeloid progenitors or colony-forming unit follow the path of differentiation (asymmetric division) or
granulocyte, erythrocyte, monocyte, and megakaryocyte (CFU- undergo apoptosis. Many theories have been proposed to
GEMM).27,28 These cells are capable of giving rise to multiple describe the mechanisms that determine the fate of the stem
lineages of blood cells. cell. Till and McCulloch proposed that hematopoiesis is a
Morphologically unrecognizable hematopoietic progenitor random process whereby the HSCs randomly commits to
cells can be divided into two major types: (1) noncommitted self-renewal or differentiation.27 This model is also called the
or undifferentiated stem cells, and (2) multipotential and com- stochastic model of hematopoiesis. Later studies suggested
mitted progenitor cells. These two groups give rise to all of the that the microenvironment in the bone marrow determines
mature blood cells. Originally there were two theories describ- whether the stem cell will self-renew or differentiate (instructive
ing the origin of hematopoietic progenitor cells. The monophy model of hematopoiesis).29 Current thinking is that the ulti-
letic theory suggests that all blood cells are derived from a single mate decision made by the stem cell can be described by both
progenitor stem cell called a pluripotential stem cell. The polyphy the stochastic and instructive models of hematopoiesis. The
letic theory suggests that each of the blood cell lineages is initial decision to self-renew or differentiate is probably sto-
derived from its own unique stem cell. The monophyletic chastic, whereas lineage differentiation that occurs later is
theory is the most widely accepted theory among experimental determined by various signals from the hematopoietic induc-
hematologists today. tive microenvironment in response to specific requirements of
Stem cells by definition can be characterized as follows: the body.
(1) they are capable of self-renewal, (2) they give rise to The multilineage priming model suggests that HSCs receive
differentiated progeny, and (3) they are able to reconstitute signals from the hematopoietic inductive microenvironment to
the hematopoietic system of a lethally irradiated host. The amplify or repress genes associated with commitment to mul-
undifferentiated HSCs can differentiate into progenitor cells tiple lineages that are expressed only at low levels. The implica-
committed to either lymphoid or myeloid lineages. These tion is that the cells fate is determined by intrinsic and extrinsic
lineage-specific progenitor cells consist of (1) the common lym factors. Extrinsic regulation involves proliferation and differen-
phoid progenitor, which proliferates and differentiates into lym- tiation signals from specialized niches located in the hemato-
phocytes of T, B, and natural killer lineages; and (2) the common poietic inductive microenvironment via direct cell-to-cell
myeloid progenitor, which proliferates and differentiates into or cellular-extracellular signaling molecules.29 Some of the
individual granulocytic, erythrocytic, monocytic, and mega- cytokines released from the hematopoietic inductive micro
karyocytic lineages. The resulting limited lineage-specific pre- environment include factors that regulate proliferation and
cursors give rise to morphologically recognizable, lineage-specific differentiation, such as stem cell factor (SCF), thrombopoietin
precursor cells (Figure 7-13 and Table 7-1). Despite the limited (TPO), and Flt3 ligand. Intrinsic regulation involves genes
numbers of HSCs in the bone marrow, more than 1 to 5 109 such as SCL (TAL1), which is expressed in cells in the heman
each of erythrocytes and leukocytes are produced each hour for gioblast, a bipotential progenitor cell of mesodermal origin
the entire life span of an individual.15 Most of the cells in that gives rise to hematopoietic and endothelial lineages,
CHAPTER 7 Hematopoiesis 77

Long-term self- Short-term self-


renewing stem cell renewing stem cell

Multipotent progenitor
hematopoietic stem cell

Common Common
myeloid progenitor lymphoid progenitor

Granulocyte-monocyte Eosinophil-basophil Megakaryocyte-erythrocyte Dendritic Pre-B Pre-T Natural


progenitor progenitor progenitor cell killer cell

Myeloblast Monoblast Myeloblast Myeloblast Pronormoblast Megakaryoblast B lymphoblast T lymphoblast

Neutrophil Monocyte Eosinophil Basophil Erythrocyte Megakaryocyte B cell T cell

Macrophage Mast cell Platelets Plasma cell


Figure 7-13 Diagram of hematopoiesis shows derivation of cells from the multipotent stem cell.

and GATA2, which is expressed in later-appearing HSCs. Both (1) decrease in basophilia, (2) increase in the proportion of
of these genes are essential for primitive and definitive hema- cytoplasm, and (3) possible appearance of granules in the
topoiesis.29 In addition to factors involved in differentiation cytoplasm. Specific changes in each lineage are discussed in
and regulation, there are regulatory signaling factors, such subsequent chapters.
as Notch-1 and Notch-2, which allow HSCs to respond to
hematopoietic inductive microenvironment factors, altering Stem Cell Cycle Kinetics
cell fate.30 The bone marrow is estimated to be capable of producing
As hematopoietic cells differentiate, they take on various approximately 3 billion erythrocytes, 2.5 billion platelets, and
morphologic features associated with maturation. These 1.5 billion granulocytes per kilogram of body weight daily. The
include an overall decrease in cell size and a decrease in the determining factor controlling the rate of production is physio
ratio of nucleus to cytoplasm. Additional changes that take logic need. Stem cells exist in the marrow in the ratio of 1 per
place during maturation occur in the cytoplasm and nucleus. 1000 nucleated blood cells. Stem cells are capable of many
Changes in the nucleus include (1) loss of nucleoli, (2) decrease mitotic divisions when stimulated by appropriate cytokines.
in the size of the nucleus, (3) condensation of chromatin, (4) When mitosis has occurred, the cell may reenter the cycle or
possible change in the shape of the nucleus, and (5) possible go into a resting phase, termed G0. Some cells in the resting
loss of the nucleus. Changes occurring in the cytoplasm include phase reenter the active cell cycle and divide one additional
78 PART II Hematopoiesis

potential colony-forming cell and the long-term colony initiating


cell, also have been identified. These hematopoietic precursor
cells give rise to colonies that can survive for 5 to 8 weeks and
be replated.30 In vivo functional assays also are available and
require transplantation of cells into syngeneic, lethally irradi-
G1
ated animals followed by transference of the engrafted bone
marrow cells into a secondary recipient.30 These systems allow
the ability of the stem cells to proliferate and differentiate to
G0 be characterized and may serve as models for developing clini-
M cally applicable techniques for gene therapy and stem cell
S transplantation.
G2
Cytokines and Growth Factors
A group of specific glycoproteins called hematopoietic growth
factors or cytokines regulates the proliferation, differentiation,
and maturation of hematopoietic precursor cells. Cytokines
are a diverse group of soluble proteins that have direct and
Figure 7-14 Cell cycle schematic. G0, Resting stage; G1, RNA and protein indirect effects on hematopoietic cells. Classification of cyto-
synthesis; S, DNA synthesis; G2, premitotic phase; M, mitosis. kines has been difficult because of their overlapping and
redundant properties. The terms cytokine and growth factor
are often used synonymously; cytokines include interleukins
time, whereas other cells are directed to terminal differentia- (ILs), lymphokines, monokines, interferons, chemokines, and colony-
tion (Figure 7-14). stimulating factors (CSFs).32 Cytokines are responsible for
From these data, a mitotic index can be calculated to stimulation or inhibition of production, differentiation, and
establish the percentage of cells in mitosis in relation to trafficking of mature blood cells and their precursors.33 Many
the total number of cells. Factors affecting the mitotic index of these cytokines exert a positive influence on stem cells
include the duration of mitosis and the length of the resting and progenitor cells with multilineage potential (e.g., IL-1,
state. Normally, the mitotic index is approximately 1% to IL-3, IL-6, IL-9, IL-11, GM-CSF, and kit ligand).33 Cytokines
2%. An increased mitotic index implies increased prolifera- that exert a negative influence on hematopoiesis include trans-
tion. An exception to this rule is in the case of megaloblastic forming growth factor-, tumor necrosis factor-, and the
anemia, in which mitosis is prolonged.31 An understanding interferons.30
of the mechanism of the generative cycle aids in under Hematopoietic precursor cells require growth factors on a
standing the mode of action of specific drugs used in the continual basis for their growth and survival. Growth factors
treatment and maintenance management of proliferative prevent hematopoietic precursor cells from dying by inhibiting
disorders. apoptosis (or programmed cell death); they stimulate them to
divide by decreasing the transit time from G0 to G1 of the cell
Stem Cell Phenotypic and Functional cycle; and they regulate cell differentiation into the various cell
Characterization lineages.
The identification and origin of stem cells can be determined Apoptosis refers to programmed cell death, a normal physi-
by immunophenotypic analysis using flow cytometry. The ologic process that eliminates unwanted, abnormal, or harmful
earliest identifiable human HSCs capable of initiating long- cells. Apoptosis differs from necrosis, which is accidental death
term cultures are CD34+, CD38, HLA-DRlow, Thy1low, and from trauma. When cells do not receive the appropriate cyto-
Lin.30 This population of marrow cells is enriched in primitive kines necessary to prevent cell death, apoptosis is initiated. In
progenitors. The expression of CD38 and HLA-DR is associated some disease states, apoptosis is turned on, which results in
with a loss of stemness. The acquisition of CD33 and early cell death, whereas in others the mechanisms involved in
CD38 is seen on committed myeloid progenitors,30 and apoptosis fail to initiate this process, which allows uncon-
the expression of CD10 and CD38 is seen on committed trolled proliferation of cells.33,34
lymphoid progenitors.30 The expression of CD7 is seen on Research techniques have accomplished the purification of
T-lymphoid progenitor cells and natural killer cells, and the many of these cytokines and the cloning of pure recombinant
expression of CD19 is seen on B-lymphoid progenitors (see growth factors, some of which are discussed in detail in the
Chapter 33).30 appropriate section of this chapter. The number of growth
Functional characterization of HSCs can be accomplished factors recognized has expanded greatly in recent years and will
through in vitro techniques using long-term culture assays. further increase as research continues. This chapter focuses
These involve the enumeration of colony-forming units (e.g., primarily on CSFs, kit ligand, Flt3 ligand, IL-3, and IL-6. A
CFU-GEMM, colony-forming unitgranulocyte-monocyte, burst- detailed discussion is beyond the scope of this text, and the
forming uniterythroid) on semisolid media, such as methyl- reader is encouraged to consult current literature for further
cellulose. Primitive progenitor cells, such as the high proliferative details.
CHAPTER 7 Hematopoiesis 79
Colony-Stimulating Factors more detail in subsequent chapters. Characteristics shared by
CSFs are produced by many different cells. They have a high interleukins include the following:
specificity for their target cells and are active at low concentra- 1. They are proteins that exhibit multiple biologic activities,
tions.32 The names of the individual factors indicate the pre- such as the regulation of autoimmune and inflammatory
dominant cell lines that respond to their presence: The primary reactions and hematopoiesis.
target of G-CSF is the granulocyte, and GM-CSF targets the 2. They have synergistic interactions with other cytokines and
granulocyte-monocyte cell line. The biologic activity of CSFs growth factors.
was first identified by their ability to induce hematopoietic 3. They are part of interacting systems with amplification
colony formation in semisolid media. In addition, it was potential.
shown in cell culture experiments that although a particular 4. They are effective at very low concentrations.
CSF may show specificity for one cell lineage, it is often capable Table 7-2 lists the sources of cytokines and sites of
of influencing other cell lineages as well. This is particularly activity.
true when multiple growth factors are combined.35 Although
G-CSF stimulates the proliferation of granulocyte progenitors,
it also works synergistically with IL-3 to enhance megakaryo-
LINEAGE-SPECIFIC HEMATOPOIESIS
cyte colony formation.35
Erythropoiesis
Erythropoiesis occurs in the bone marrow and is a complex,
Stem Cell Factor, Flt3 Ligand, regulated process for maintaining adequate numbers of eryth-
Granulocyte-Monocyte Colony-Stimulating rocytes in the peripheral blood. The CFU-GEMM gives rise to
Factor, Interleukin-3, and Interleukin-6 the earliest identifiable colony of RBCs, called the burst-forming
Ogawa36 described early-acting growth factors (multilineage), uniterythroid (BFU-E). The BFU-E produces a large multiclus-
intermediate-acting growth factors (multilineage), and late- tered colony that resembles a cluster of grapes containing
acting growth factors (lineage restricted). Kit ligand, also brightly colored hemoglobin. These colonies range from a
known as SCF, is an early-acting growth factor; its cell surface single large cluster to 16 or more clusters. BFU-Es contain only
receptor is the product of the c-kit gene. The receptor c-kit is a a few receptors for EPO, and their cell cycle activity is not
type I tyrosine kinase receptor that is expressed on HSCs and is influenced significantly by the presence of exogenous EPO.
downregulated with differentiation. The binding of SCF to its BFU-Es under the influence of IL-3, GM-CSF, TPO, and kit
receptor (c-kit) induces a series of signals that are sent via signal ligand develop into colony-forming uniterythroid (CFU-E)
transduction pathways from the cytoplasmic receptor of the colonies.28 The CFU-E has many EPO receptors and has an
HSC to the nucleus of the HSC, stimulating the cell to prolifer- absolute requirement for EPO. Some CFU-Es are responsive to
ate. As HSCs differentiate and mature, the expression of c-kit low levels of EPO and do not have the proliferative capacity of
decreases. Activation of kit ligand by SCF is essential in the the BFU-E.42
early stages of hematopoiesis.33,37 EPO is a lineage-specific glycoprotein that prevents apopto-
Flt3 seems to act at an even earlier stage of HSC develop- sis of erythroid precursors. EPO is produced in the renal peri
ment than SCF. Flt3 stands for c-fms-like tyrosine kinase.38 SCF tubular interstitial cells or renal tubular cells.42 In addition, a
and Flt3 work synergistically with IL-3, G-CSF, and GM-CSF to small amount of EPO is produced by the liver.37 EPO induces
promote early HSC proliferation. hemoglobin synthesis and serves as a differentiation factor
Other early-acting CSFs that are multilineage include IL-3, causing the CFU-E to differentiate into pronormoblasts, the
GM-CSF, and IL-6. IL-3 regulates blood cell production by earliest visually recognized erythrocyte precursors in the bone
controlling the production, differentiation, and function of marrow.43 Erythropoiesis is discussed in detail in Chapter 8.
granulocytes and macrophages.39 GM-CSF induces expression
of specific genes and stimulates HSC proliferation, differentia-
tion, and functional activation.40 IL-6 is a pleiotropic growth Leukopoiesis
factor with stimulatory effects on myeloid and lymphoid cell Leukopoiesis can be divided into two major categories: myelo
lineages.41 poiesis and lymphopoiesis. Factors that promote differentiation
of the CFU-GEMM into neutrophils, monocytes, eosinophils,
and basophils include GM-CSF, G-CSF, monocyte colony-
Interleukins stimulating factor (M-CSF), IL-3, IL-5, IL-11, and kit ligand.
Cytokines originally were named according to their specific GM-CSF stimulates the proliferation and differentiation of
function, such as lymphocyte-activating factor (now called IL- neutrophil and macrophage colonies from the colony-forming
1), but continued research showed that a particular cytokine unitgranulocyte-monocyte. G-CSF and M-CSF stimulate neu-
may have multiple actions. A group of scientists began calling trophil differentiation and monocyte differentiation from the
some of the cytokines interleukins, numbering them in the colony-forming unitgranulocyte and colony-forming unit
order in which they were identified (e.g., IL-1, IL-2). Figure monocyte.42 IL-3 is a multilineage stimulating factor that stim-
7-15 illustrates the hematopoietic system and the sites of ulates the growth of granulocytes, monocytes, megakaryocytes,
action of some of the cytokines. These factors are discussed in and erythroid cells. Eosinophils require GM-CSF, IL-5, and IL-3
80 PART II Hematopoiesis

Long-term self- Short-term self-


renewing stem cell renewing stem cell

SCF, IL-1, 3, 6

Multipotent progenitor
hematopoietic stem cell

FL, SCF, GM-CSF FL, SCF


IL-1, 3, 6, 11 IL-7

Common Common
myeloid progenitor lymphoid progenitor
FL, SCF, GM-CSF SCF, GM-CSF SCF
SCF, GM-CSF
IL-3 IL-3, 5 IL-3 FL FL FL, IL-7
IL-7 IL-7

Granulocyte-monocyte Eosinophil-basophil Megakaryocyte-erythrocyte Dendritic Pre-B Pre-T Natural


progenitor progenitor progenitor cell killer cell

G-CSF M-CSF EPO TPO

Myeloblast Monoblast Myeloblast Myeloblast Pronormoblast Megakaryoblast B lymphoblast T lymphoblast

Neutrophil Monocyte Eosinophil Basophil Erythrocyte Megakaryocyte B cell T cell

Macrophage Mast cell Platelets Plasma cell


Figure 7-15 Diagram of derivation of hematopoietic cells, illustrating sites of activity of CSFs and ILs. EPO, Erythropoietin; FL, Flt ligand; G-CSF, granulocyte
colony-stimulating factor; GM-CSF, granulocyte-monocyte colony-stimulating factor; M-CSF, monocyte colony-stimulating factor; SCF, stem cell factor;
TPO, thrombopoietin; IL-1, interleukin-1; IL-3, interleukin-3; IL-5, interleukin-5; IL-6, interleukin-6; IL-7, interleukin-7; IL-11, interleukin-11.

for differentiation. The requirements for basophil differentia-


ANALYTICAL AND
tion are less clear, but it seems to depend on the presence of
THERAPEUTIC APPLICATIONS
IL-3 and kit ligand. Growth factors promoting lymphoid dif-
ferentiation include IL-2, IL-7, IL-12, and IL-15 and to some Clinical use of growth factors approved by the U.S. Food and
extent IL-4, IL-10, IL-13, IL-14, and IL-16.38 Leukopoiesis is Drug Administration has contributed numerous options in the
discussed further in Chapter 12. treatment of hematologic malignancies and solid tumors. In
addition, growth factors can be used as priming agents to
Megakaryopoiesis increase the yield of HSCs during apheresis for transplantation
Earlier influences on megakaryopoiesis include GM-CSF, IL-3, protocols. Advances in molecular biology have resulted in
IL-6, IL-11, kit ligand, and TPO.34 The stimulating hormonal cloning of the genes that are responsible for the synthesis of
factor TPO (also known as mpl ligand), along with IL-11, con- various growth factors and the recombinant production of
trols the production and release of platelets. The liver is the large quantities of these proteins. Table 7-2 is a concise over-
main site of production of TPO.44,45 Megakaryopoiesis is dis- view of some growth factors, physiologic roles, and applica-
cussed in Chapter 13. tions. Many more examples can be found in the literature.
CHAPTER 7 Hematopoiesis 81

TABLE 7-2 Selected Cytokines, Sources, Target, and Clinical Application


Cytokine Source Target Clinical Application
IL-1 Monocytes/macrophages, endothelial Fibroblasts, endothelial cells, Used in cases of inflammation, such as fever, and as a
cells, fibroblasts, epithelial cells, T basophils defense mechanism during infection43,46
cells
IL-2 T cells T, B, NK cells Enhances migration of cells with antitumor activity into
tissues; induces release of IL-6, tumor necrosis
factor, and interferon, which may have antitumor
effects47
IL-3 T cells, monocytes/macrophages, mast Macrophages, neutrophils, Used in treatment of refractory anemia with and without
cells eosinophils, basophils, mast cells, ringed sideroblasts; also used to stimulate
megakaryocytes, T and B cells megakaryopoiesis48
IL-4 T cells, B cells, macrophages, Macrophages, myeloid progenitors, T Used to potentiate antitumor activity, especially in cases
basophils, mast cells and B cells, mast cells, fibroblasts of gastric carcinoma49
IL-5 T cells T and B cells, eosinophils
IL-6 Monocytes/macrophages, endothelial Fibroblasts, myeloid precursors, T Pleiotropic cytokine with wide range of biologic
cells, T cells, fibroblasts, osteoblasts and B cells, mast cells, fibroblasts activities in immune regulation and hematopoiesis
IL-7 BM stromal cells, spleen, thymus Pre-B cells, early T cells, CLL, Szary Enhances allocytolytic activity of lymphokine-activated
cells, ALL killer cells50; is considered to be a promoter of
thymopoiesis and immune recovery51
IL-8 Monocytes/macrophages, fibroblasts, Chemoattractant for neutrophils, Active in neutrophil chemotaxis and degranulation; acts
neutrophils, endothelial cells, T cells, eosinophils as a principal mediator of inflammation that then can
synovial cells, chondrocytes be acted on by IL-452
IL-9 T cells T cells, macrophages, mast cells Potent lymphoid cell factor, stimulating growth of
certain T cells and mast cells53,54
IL-10 T and B cells, macrophages Lymphoid and myeloid cells Synergistic activity of IL-4 and IL-10 causes
antiinflammatory effect on human placental cells
in vitro55
IL-11 BM stromal cells, fibroblasts Progenitor cells and stromal cells Has shown activity in preclinical studies in association
with myelosuppression, cancer therapy, neutropenia
and thrombocytopenia56; stimulates growth and
survivability of certain B and T cells57
IL-12 Macrophages T, NK cells Used in stimulating proliferation of peripheral blood
mononuclear cells and tumor-infiltrating lymphocytes
in melanoma patients57; capable of reversing
immunosuppression mediated by the neoplastic
agent paclitaxel58
IL-13 T cells, basophils, stromal cells Monocytes, B lymphocytes Pleiotropic cytokine that has important antiinflammatory
and immunoregulatory activities50
IL-14 T cells, T and B lymphoma cells Pre-B, T, NK cells CD4+ cell growth factor; proinflammatory; enhances
lymphocyte chemotaxis59
IL-15 Monocytes/macrophages, epithelial T, B, and NK cells, mast cells rhIL-15 has clinical applications in immunodeficiency
cells, skeletal muscle cells, BM and diseases and BM transplantation, where it might be
thymic stromal cells advantageous to accelerate peripheral expansion and
promote localization of T cells60
IL-16 CD8+ T cells, mast cells T and B cells Chemoattracts CD4 T cells61
IL-17 CD4+ cells Macrophages May be useful in further delineating mechanisms of
G-CSF regulation50
IL-18 Monocytes/macrophages, liver Tumor cells, pathogens
G-CSF Endothelial cells, placenta, monocytes/ All granulocytes, macrophages, Stimulates granulocyte production and granulocyte
macrophages, fibroblasts endothelial cells, fibroblasts, activation62; may be used to treat neutropenia and to
leukemic myeloblasts mobilize CD34+ stem cells in the peripheral blood63
M-CSF B and T cells, endothelial cells, Monocytes/macrophages, osteoblasts Used to stimulate monocytes/macrophages production
monocytes/macrophages, fibroblasts, and activity62
epithelial cells, osteoblasts
continued
82 PART II Hematopoiesis

TABLE 7-2 Selected Cytokines, Sources, Target, and Clinical Applicationcontd


Cytokine Source Target Clinical Application
GM-CSF B, T, and NK cells, osteoblasts, All granulocytes, megakaryocytes, May be used to treat neutropenia in patients receiving
endothelial cells, fibroblasts, stem cells, BFU-E, monocytes/ chemotherapy and in those undergoing BM
monocytes/macrophages macrophages, leukemic transplantation64
myeloblasts
EPO Kidney, liver BFU-E, CFU-E Used to stimulate growth and proliferation of erythroid
precursors; used clinically to treat anemia associated
with renal failure, chemotherapy, and BM infiltration
by cancer64
TPO Kidney, liver, spleen, BM stroma, Megakaryocytes, platelets, c-mpl+ Essential for maturation of megakaryocytes and
muscle, brain blasts enhances platelet production; used to speed
recovery of blood platelets after cytoreductive
therapies65
c-kit ligand Fibroblasts, BM stroma, hepatocytes Primitive progenitors Also referred to as stem cell factor/steel factor; used to
stimulate myeloid, erythroid, and lymphoid
progenitors66
Flt3 ligand BM fibroblasts and stroma cells Acts synergistically with other growth Stimulates primitive progenitor cells68
factors to stimulate T cells, B
cells, NK cells, and dendritic
cells67

ALL, Acute lymphocytic leukemia; BFU-E, burst-forming uniterythroid; BM, bone marrow; CFU-E, colony-forming uniterythroid; CLL, chronic lymphocytic leukemia;
EPO, erythropoietin; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-monocyte colony-stimulating factor; IL, interleukin; M-CSF, monocyte colony-stimulating
factor; mono/macro, monocytes/macrophages; NK, natural killer; rhIL, recombinant human interleukin; TPO, thrombopoietin.

SUMMARY
Hematopoiesis is the formation, development, and specialization of maturation include decrease in cell size, decrease in nuclear
of all functional blood cells. size, loss of nucleoli, condensation of nuclear chromatin, and
Hematopoiesis progresses through the mesoblastic, hepatic, and decreased basophilia in cytoplasm. Some morphologic changes
medullary phases. are unique to specific lineages (e.g., loss of the nucleus in
Organs that function at some point in hematopoiesis include the RBCs).
liver, spleen, lymph nodes, thymus, and bone marrow. Cytokines and growth factors play a major role in determining
The bone marrow is the primary site of hematopoiesis at birth and differentiation of stem cells.
throughout life. Cytokines include ILs, CSFs, interferons, and others.
In certain situations, blood cell production may occur outside the Some cytokines exert influence on stem cells with multilineage
bone marrow; such production is termed extramedullary. potential, some are lineage specific, and some function only in
The microenvironment in the bone marrow is essential for stem combination with other cytokines.
cell differentiation and proliferation. Cytokines are necessary to prevent premature apoptosis.
Monophyletic theory suggests that all blood cells arise from a Cytokines have contributed new options in the treatment of bone
common progenitor. marrow malignancies, leukemias, and aplastic anemias.
As cells mature, certain morphologic characteristics of maturation
allow specific lineages to be recognized. General characteristics

R E V I E W Q UESTIONS
1. The process of formation and development of blood cells 2. During midfetal life, the primary source of blood cells is
is termed: the:
a. Hematopoiesis a. Bone marrow
b. Hematemesis b. Spleen
c. Hematocytometry c. Lymph nodes
d. Hematorrhea d. Liver
CHAPTER 7 Hematopoiesis 83

3. Which of the following organs is responsible for the con- 7. The term for the earliest hematopoietic cell is:
ditioning of T lymphocytes? a. Prehematopoietic blast
a. Spleen b. Pluripotential stem cell
b. Liver c. CFU-GEMM
c. Thymus d. Cytokinetic precursor
d. Bone marrow
8. Which of the following cells is not a product of the
4. The best source of active bone marrow from a 20-year-old CFU-GEMM?
would be: a. Megakaryocyte
a. Iliac crest (hip) b. Lymphocyte
b. Femur (thigh) c. Erythrocyte
c. Distal radius (forearm) d. Granulocyte
d. Tibia (shin)
9. Which of the following hematopoietic growth factors is
5. Physiologic programmed cell death is termed: produced in the kidney?
a. Angiogenesis a. EPO
b. Apoptosis b. TPO
c. Aneurysm c. GM-CSF
d. Apohematics d. M-CSF

6. Which organ is the site of sequestration of platelets? 10. A multilineage cytokine among the ILs is:
a. Liver a. IL-1
b. Thymus b. IL-2
c. Spleen c. IL-3
d. Bone marrow d. IL-4

REFERENCES
1. Marcos MA, Godlin I, Cumano A, et al: Developmental events 12. Warren J, Ward P: The inflammatory response. In Beutler E,
from hemopoietic stem cells to B-cell populations and Ig Lichtman M, Coller B, et al, editors: Williams hematology,
repertoires. Immunol Rev 137:155-171, 1994. ed 7, New York, 2006, McGraw-Hill, pp 221-230.
2. Schoemans H, Verfaillie CM: Cellular biology of hemato 13. Abboud C, Lichtman MA: Structure of the marrow and the
poiesis. In Hoffman R, Benz EJ, Shattil SJ, et al, editors: hematopoietic environment. In Beutler E, Lichtman M,
Hematology basic principles and practice, ed 5, New York, 2009, Coller B, et al, editors: Williams hematology, ed 7, New York,
Churchill Livingstone, pp 200-212. 2006, McGraw-Hill, pp 35-72.
3. Peault B: Hematopoietic stem cell emergence in embryonic 14. Schlueter AI: Structure and function of hematopoietic organs.
life: developmental hematology revisited. J Hematotherapy In McKenzie SB, editor: Clinical laboratory hematology, ed 2,
5:369-378, 1996. Upper Saddle River, NJ, 2010, Prentice Hall, pp 51-61.
4. Tavian M, Coulombel L, Luton D, et al: Aorta-associated 15. Gupta P, Blazar B, Gupta K, et al: Human CD34+ bone
CD34+ hematopoietic cells in the early human embryo. Blood marrow cells regulate stromal production of interleukin-6
87:67-72, 1996. and granulocyte colony-stimulating factor and increase the
5. Charbord P, Tavian M, Coulombel L, et al: Early ontogeny of colony-stimulating activity of stroma. Blood 91:3724-3733,
the human hematopoietic system [in French], C R Seances Soc 1998.
Biol Fil 189:601-609 (abstract), 1995. 16. Klein G: The extracellular matrix of the hematopoietic micro-
6. Dieterlen-Lievre F, Godin I, Pardanaud I: Where do hemato- environment. Experientia 51:914-926, 1995.
poietic stem cells come from? Arch Allergy Immunol 112:3-8, 17. Sadahira Y, Mori M: Role of the macrophage in erythropoi-
1997. esis. Pathol Int 10:841-848, 1999.
7. Chang Y, Paige CJ, Wu GE: Enumeration and characterization 18. Thibodeau GA, Patton KT: Anatomy and physiology, ed 5,
of DJH structures in mouse fetal liver. EMBO J 11:1891-1899, St Louis, 2003, Mosby.
1992. 19. Weinberg JB: Mononuclear phagocytes. In Greer JP, Foerster
8. Gallicchio VS: Hematopoiesis and review of genetics. In J, Lukens J, et al, editors: Wintrobes clinical hematology,
Steine-Martin EA, Lotspeich-Steininger CA, Koepke JA, ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott
editors: Clinical hematology: principles, procedures, correlations, Williams & Wilkins, pp 249-281.
ed 2, Philadelphia, 1998, JB Lippincott, pp 46-56. 20. Warkentin TE, Kelton JG: Thrombocytopenia due to platelet
9. Mescher AL: Junqueiras basic histology, ed 12, Stamford, Conn, destruction and hypersplenism. In Hoffman R, Benz EJ,
2010, Appleton & Lange. Shattil SJ, et al, editors: Hematology basic principles and
10. Ross MH, Pawlina W: Histology: a text and atlas: with correlated practice, ed 5, New York, 2009, Churchill Livingstone,
cell and molecular biology, ed 5, Philadelphia, 2006, Lippincott pp 2113-2132.
Williams & Wilkins, pp 247-279. 21. Seeley RR, Stephens D, Tate P: Anatomy and physiology, ed 3,
11. Segel G, Palis J: Hematology of the newborn. In Beutler E, St Louis, 1995, Mosby.
Lichtman M, Coller B, et al, editors: Williams hematology, 22. Shurin SB: The spleen and its disorders. In Hoffman R,
ed 7, New York, 2006, McGraw-Hill, pp 81-99. Benz EJ, Shattil SJ, et al, editors: Hematology basic principles
84 PART II Hematopoiesis

and practice, ed 5, New York, 2009, Churchill Livingstone, 41. Kopf M, Ramsay A, Brombacher F, et al: Peliotropic defects of
pp 2419-2429. IL-6 deficient mice including early hematopoiesis, T and B
23. Ware RE: The autoimmune hemolytic anemias. In Nathan cell function, and acute-phase responses. Ann N Y Acad Sci
DG, Orkin SH, editors: Nathan and Oskis hematology of 762:308-318, 1995.
infancy and childhood, ed 7, Philadelphia, 2009, Saunders, 42. Kaushansky K: Hematopoietic stem cells, progenitor
pp 613-658. cells, and cytokines. In Beutler E, Lichtman MA, Coller BS,
24. Porembka MR, Majella Doyl MB, Chapman WC: Disorders et al, editors: Williams hematology, ed 7, New York, 2009,
of the spleen. In Greer JP, Foerster J, Lukens J, et al, editors: McGraw-Hill, pp 201-220.
Wintrobes clinical hematology, ed 12, Philadelphia, 2009, 43. Sawada K, Krantz SB, Dai CH: Purification of human burst-
Wolters Kluwer Health/Lippincott Williams & Wilkins, forming units-erythroid and demonstration of the evolution
pp 1637-1654. of erythropoietin receptors. J Cell Physiol 142:219-230,
25. Kipps TJ: The lymphoid tissues. In Beutler E, Lichtman M, 1990.
Coller B, et al, editors: Williams hematology, ed 7, New York, 44. Wolber EM, Ganschow R, Burdelski M, et al: Hepatic throm-
2006, McGraw-Hill, pp 73-80. bopoietin mRNA levels in acute and chronic liver failure of
26. Paraskevas F: T lymphocytes and NK cells. In Greer JP, childhood. Hepatology 29:1739-1742, 1999.
Foerster J, Lukens J, et al, editors: Wintrobes clinical hemato 45. Wolber EM, Dame C, Fahnenstich H, et al: Expression of the
logy, ed 12, Philadelphia, 2009, Wolters Kluwer Health/ thrombopoietin gene in human fetal and neonatal tissues.
Lippincott Williams & Wilkins, pp 358-401. Blood 94:97-105, 1999.
27. Till TE, McCulloch EA: A direct measurement of the radiation 46. Scales WE: Structure and function of interleukin-1. In Kunkel
sensitivity of normal mouse marrow cells. Radiat Res 14:213- SL, Remick DG, editors: Cytokines in health and disease.
222, 1961. New York, 1992, Marcel Dekker, pp 15-26.
28. Dessypris EN, Sawyer ST: Erythropoiesis. In Greer JP, Foerster 47. Rosenberg SA, Lotze MT, Muul LM, et al: A progress report of
J, Lukens J, et al, editors: Wintrobes clinical hematology, the treatment of 157 patients with advanced cancer using
ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott lymphokine-activated cells and IL-2 or high dose IL-2 alone.
Williams & Wilkins, pp 106-155. N Engl J Med 316:889-987, 1987.
29. Metcalf D: On hematopoietic stem cell fate. Immunity 26:669- 48. Vrhovac R, Kusee R, Jaksic B: Myeloid hematopoietic growth
673, 2007. factor. Int J Clin Pharmacol 31:241-252, 1988.
30. Verfaillie C: Regulation of hematopoiesis. In Wickramasinghe 49. Morisake T, Yuzuki D, Lin R, et al: Interleukin-4 receptor
SN, McCullough J, editors: Blood and bone marrow pathology. expression and growth inhibition of gastric carcinoma cells
New York, 2003, Churchill Livingstone, pp 71-85. by interleukin-4. Cancer Res 52:6059-6060, 1991.
31. Antony A: Megaloblastic anemias. In Hoffman R, Benz EJ, 50. deWall-Malefyt R, Figdor CG, Huijbens R, et al: Effects of
Shattil SJ, et al, editors: Hematology basic principles and IL-13 on phenotype, cytokine production, and cytotoxic
practice, ed 4, New York, 2009, Churchill Livingstone, function of human monocytes. J Immunol 151:6370-6381,
pp 491-524. 1993.
32. COPE: Cytokines Online Pathfinder Encyclopaedia, Horst 51. Malave I, Vethencourt MA, Chacon R, et al: Production of
Ibelfaufts Hypertext Information Universe of Cytokines, interleukin-6 in cultures of peripheral blood mononuclear
version 4.0, August 1999. Available at: http://www. cells from children with primary protein-calorie malnutrition
copewithcytokines.de. Accessed October 21, 2009. and from eutrophic controls. Ann Nutr Metab 42:266-273,
33. Mathurs S, Schexneider K, Hutchinson RE: Hematopoiesis. 1998.
In Henry JB, editor: Clinical diagnosis and management by 52. Schroder IM: Peptides and cytokines. Arch Dematol Res
laboratory methods, ed 21, Philadelphia, 2007, Saunders, 284(suppl 1):S22-S26, 1992.
pp 484-503. 53. Michaels LA, Ohene-Fremepong K, Zhao H, et al: Serum
34. Shaheen M, Broxmeyer HE: The humoral regulation of hema- levels of substance P are elevated in patients with sickle cell
topoiesis. In Hoffman R, Benz EJ, Shattil SJ, et al, editors: disease and increase further during vaso-occlusive crisis. Blood
Hematology basic principles and practice, ed 5, New York, 2009, 92:3148-3151, 1998.
Churchill Livingstone, pp 253-275. 54. Modi WS, Pollack DD, Mock BA, et al: Regional localization
35. Sieff C, Zon LI: The anatomy and physiology of hemato of the human glutaminase (GLS) and interleukin-9 (IL-9)
poiesis. In Nathan DG, Orkin SH, editors: Nathan and Oskis genes by in situ hybridization. Cytogenet Cell Genet 57:114-
hematology of infancy and childhood, ed 7, Philadelphia, 2009, 116, 1991.
Saunders, pp 195-273. 55. Goodwin VJ, Sato TA, Mitchell MD, et al: Anti-inflammatory
36. Ogawa M: Differentiation and proliferation of hematopoietic effects of IL-4 and IL-10 and transforming growth factor-beta
stem cells. Blood 81:2844-2853, 1993. on human placental cells in vitro. Am J Reprod Immunol
37. Koury MJ, Mahmud N, Rhodes M: Origin and development 40:319-325, 1998.
of blood cells. In Greer JP, Foerster J, Lukens J, et al, editors: 56. Neben S, Tumer K: The biology of interleukin-11. Stem Cells
Wintrobes clinical hematology, ed 12, Philadelphia, 2009, 11(suppl 2):156-162, 1993.
Wolters Kluwer Health/Lippincott Williams & Wilkins, 57. Zeh HJ, Hurd S, Strojus WJ, et al: Interleukin-12 promotes
pp 79-105. the proliferation and cytolytic maturation of immune
38. Shaheen M, Broxmeyer H: The humoral regulation of hema- effectors: implications for the immunotherapy of cancer.
topoiesis. In Hoffman R, Benz EJ, Shattil SJ, et al, editors: J Immunother 14:155-161, 1993.
Hematology basic principles and practice, ed 5, New York, 2009, 58. Mullins DW, Koci MD, Burger CJ, et al: Interleukin-12 over-
Churchill Livingstone, pp 253-275. comes paclitaxel-mediated suppression of T-cell prolifera-
39. Dorssers L, Burger H, Bot F, et al: Characterization of a human tion. Immunopharmacol Immunotoxicol 20:473-492, 1998.
multilineage-colony-stimulating factor cDNA clone identi- 59. Leca N, Laftavi M, Shen L, et al: Regulation of human inter-
fied by a conserved noncoding sequence in mouse leukin 14 transcription in vitro and in vivo after renal trans-
interleukin-3. Gene 55:115-124, 1987. plantation. Transplantation 27;86:336-341, 2008 Jul.
40. Lin EY, Orlofsky A, Berger MS, et al: Characterization of 60. Sun A, Wei H, Sun R, et al. Human interleukin-15 improves
A1, a novel hemopoietic-specific early-response gene with engraftment of human T cells in NOD-SCID mice. Clin
sequence similarity to bcl-s. J Immunol 151:1979-1988, 1993. Vaccine Immunol 13:227-234, 2006.
CHAPTER 7 Hematopoiesis 85
61. Cruikshank WW, Kornfeld H, Center DM: Interleukin-16. 65. Kaushansky K: Thrombopoietin: a tool for understanding
J Leukoc Biol 67:757-766, 2000. thrombopoiesis. J Thromb Haemost 1:1587-1592, 2003.
62. Bagby GC, Segal, GM: Cytokines with hematopoietic activi- 66. Quesinaux VFJ: Interleukins 9, 10, 11, and 12 and kit-ligand:
ties. In Hoffman R, Berry EJ, Shattil SJ, et al, editors: Hematol a brief overview. Res Immunol 143:385-400, 1992.
ogy, basic principles and practice, ed 5, New York, 2009, 67. Karsunky H, Miriam M, Cozzio A, et al: Flt3 ligand regulates
Churchill Livingstone, p 97. dendritic cell development from Flt3+ lymphoid and-myeloid
63. Hubel K, Engert A: Clinical applications of granulocyte committed progenitors to Flt3+ dendritic cells in vivo.
colony-stimulating factor: an update and summary. Ann J Environ Monit 198:305-313, 2003.
Hematol 82:207-213, 2003. 68. Gratwohl A, John L, Baldomero H, et al: FLT-3 ligand pro-
64. Mertelsmann R: Hematopoietic cytokines: From biology vides hematopoietic protection from total body irradiation
and pathophysiology to clinical application. Leukemia in rabbits. Blood 92:765-769, 1998.
(suppl 2):S168-S177, 1993.
8 Erythrocyte Production and Destruction Kathryn Doig

OUTLINE OBJECTIVES
Normoblastic Maturation After completion of this chapter, the reader will be able to:
Terminology
Maturation Process 1. List and describe the erythroid precursors in order 11. Explain how hypoxia stimulates RBC production.
Criteria Used in of maturity, including the morphologic characteris- 12. Describe the general chemical composition of eryth-
Identification of the tics, cellular activities, normal location, and length of ropoietin (EPO) and name the site of production.
Erythroid Precursors time in the stage for each. 13. Discuss the various mechanisms by which EPO con-
Maturation Sequence 2. Correlate the erythroblast, normoblast, and rubriblast tributes to erythropoiesis.
Erythrokinetics nomenclatures for red blood cell (RBC) stages. 14. Define and explain apoptosis resulting from Fas/FasL
Hypoxiathe Stimulus to 3. Name the stage of erythroid development when interactions and how this regulatory mechanism
Red Blood Cell given a written description of the morphology of a applies to erythropoiesis.
Production cell in a Wright-stained bone marrow smear. 15. Explain the effect of bcl-xL and the general mecha-
Other Stimuli to
4. List and compare the cellular organelles of immature nism by which it is stimulated in red blood cell
Erythropoiesis
and mature erythrocytes and describe their specific progenitors.
Microenvironment of the
Bone Marrow functions. 16. Describe the features of the bone marrow that
Erythrocyte Destruction 5. Name the erythrocyte progenitors and distinguish contribute to establishing the microenvironment
Macrophage-Mediated them from precursors. necessary for the proliferation of RBCs, including
Hemolysis 6. Explain the nucleus-to-cytoplasm (N:C) ratio. location and arrangement relative to other cells, with
(Extravascular Describe the appearance of a cell when given the particular emphasis on the role of fibronectin.
Hemolysis) N:C ratio or estimate the N:C ratio from the appear- 17. Discuss the role of macrophages in RBC
Mechanical Hemolysis ance of a cell. development.
(Intravascular 7. Explain how reticulocytes can be recognized in a 18. Explain how RBCs enter the bloodstream and how
Hemolysis) Wright-stained peripheral blood film. premature entry is prevented and, when appropriate,
8. Define and differentiate the terms polychromasia, promoted.
diffuse basophilia, punctate basophilia, and baso- 19. Describe the characteristics of senescent RBCs and
philic stippling. explain why RBCs age.
9. Discuss the differences between the reticulum of 20. Explain and differentiate the two normal mechanisms
reticulocytes and punctate basophilic stippling in of erythrocyte destruction, including location and
composition and conditions for viewing. process.
10. Define and differentiate erythron and RBC mass.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 42-year-old premenopausal woman has emphysema. This cell (RBC) count to be 5.8 1012/L. After she has received
lung disease impairs the ability to oxygenate the blood, so oxygen for several months, her RBC count is 5.0 1012/L.
patients experience significant fatigue and tiredness. To allevi- 1. What explains the elevation of the first RBC count?
ate these symptoms, oxygen is typically prescribed, and this 2. What hormone is responsible? How is its production
patient has a portable oxygen tank she carries with her at all stimulated? What is the major way in which it acts?
times, breathing through nasal cannulae. Before she began 3. What explains the decline in RBC count with oxygen
using oxygen, a complete blood count showed her red blood therapy for this patient?

86
CHAPTER 8 Erythrocyte Production and Destruction 87

T
he red blood cell (RBC), or erythrocyte, provides a TABLE 8-1 Three Erythroid Precursor Nomenclature
classic example of the biologic principle that cells have Systems
specialized functions and that their structures are spe-
Nomenclature Maturation Stages
cific for those functions. The erythrocyte has one true function:
to carry oxygen from the lung to the tissues where the oxygen Normoblastic Pronormoblast
is released. This is accomplished by the attachment of the Basophilic normoblast
oxygen to hemoglobin (Hb), the major cytoplasmic compo- Polychromatic (polychromatophilic) normoblast
nent of mature RBCs. The role of the RBC in returning carbon Orthochromic normoblast
dioxide to the lungs and buffering the pH of the blood is Polychromatic (polychromatophilic) erythrocyte
important but is quite secondary to its oxygen-carrying func- Erythrocyte
tion. To protect this essential life function, the mechanisms Rubriblastic Rubriblast
controlling development, production, and normal destruction Prorubricyte
of RBCs are fine-tuned to avoid interruptions in oxygen Rubricyte
delivery, even under adverse conditions such as blood loss Metarubricyte
with hemorrhage. This chapter and subsequent chapters Polychromatic (polychromatophilic) erythrocyte
discussing iron, RBC metabolism, membrane structure, and Erythrocyte
hemoglobin constitute the foundation for understanding the Erythroblastic Proerythroblast
bodys response to diminished oxygen-carrying capacity of the Basophilic erythroblast
blood, called anemia. Polychromic (polychromatophilic) erythroblast
Although in some ways a simple cell, and thus highly Orthochromic erythroblast
studied, the mammalian erythrocyte is unique among animal Polychromatic (polychromatophilic) erythrocyte
cells in having no nucleus in its mature, functional state. While Erythrocyte
amphibians and birds possess RBCs similar to those of
mammals, the nonmammalian cells retain their nuclei through-
out the cells lives. The implications of this unique mammalian
adaptation are significant for cell function and life span. three to five divisions before maturing further.1 As seen later,
it takes approximately another 6 days for the precursors to
become mature cells ready to enter the circulation, so approxi-
NORMOBLASTIC MATURATION
mately 18 to 21 days are required to produce a mature RBC
Terminology from the BFU-E.
RBCs are formally called erythrocytes. The nucleated precursors
in the bone marrow are called erythroblasts. They also may be Erythroid Precursors
called normoblasts, which refers to developing nucleated cells Normoblastic proliferation, similar to the proliferation of
(i.e., blasts) with normal appearance. This is in contrast to the other cell lines, is a process encompassing replication (i.e.,
abnormal appearance of the developing nucleated cells in division) to increase cell numbers and development from
megaloblastic anemia, in which the erythroblasts are called immature to mature cell stages (Figure 8-1). The earliest mor-
megaloblasts because of their large size. phologically recognizable erythrocyte precursor, the pronor-
Three nomenclatures are used for naming the erythroid moblast, is derived via the BFU-E and CFU-E from the
precursors (Table 8-1). The erythroblast terminology is used multipotential stem cells as discussed in Chapter 7. The pro-
primarily in Europe. Like the normoblastic terminology used normoblast is able to divide, with each daughter cell maturing
more often in the United States, it has the advantage of being to the next stage of development, the basophilic normoblast.
descriptive of the appearance of the cells. Some prefer the Each of these cells can divide, with each of its daughter cells
rubriblast terminology because it parallels the nomenclature maturing to the next stage, the polychromatophilic normo-
used for white blood cell development. Normoblastic termi- blast. Each of these cells also can divide and mature. In the
nology is used in this chapter. erythrocyte cell line, there are typically three and occasionally
as many as five divisions2 with subsequent nuclear and cyto-
Maturation Process plasmic maturation of the daughter cells, so that from a single
Erythroid Progenitors pronormoblast, eight mature RBCs usually result. The condi-
As described in Chapter 7, the morphologically identifiable tions under which the number of divisions can be increased or
erythrocyte precursors develop from two functionally identifi- reduced are discussed later.
able progenitors, burst-forming uniterythroid (BFU-E) and
colony-forming uniterythroid (CFU-E), committed to the ery- Criteria Used in Identification of
throid cell line. Estimates of time spent at each stage suggest the Erythroid Precursors
that it takes about 1 week for the BFU-E to mature to the CFU-E Morphologic identification of blood cells depends on a well-
and another week for the CFU-E to become a pronormoblast,1 stained peripheral blood film or bone marrow smear (see
which is the first morphologically identifiable RBC precursor. Chapter 16). In hematology, a modified Romanowsky stain,
While at the CFU-E stage, the cell completes approximately such as Wright or Wright-Giemsa, is commonly used. The
88 PART II Hematopoiesis

Multipotential
stem cell

Pronormoblast

Basophilic
normoblast

Polychromatophilic
normoblast

Orthochromic
normoblast

Shift
reticulocyte BM

Reticulocyte PB

Erythrocyte

Figure 8-1 Typical production of erythrocytes from two pronormoblasts illustrating three mitotic divisions. BM, Bone marrow; PB, peripheral blood
circulation.

As RBCs mature, several general trends affect their appear-


BOX 8-1 Nucleus-to-Cytoplasm (N:C) Ratio ance. Figure 8-2 graphically represents these trends.
The nucleus-to-cytoplasm (N:C) ratio is a morphologic feature used 1. The overall diameter of the cell decreases.
to identify and stage red blood cell and white blood cell precursors. 2. The diameter of the nucleus decreases more rapidly than
The ratio is a visual estimate of what area of the cell is occupied by does the size of the cell. As a result, the N:C ratio also
the nucleus compared with the cytoplasm. If the areas of each are decreases.
approximately equal, the N:C ratio is 1:1. Although not mathemati- 3. The nuclear chromatin pattern becomes coarser, clumped,
cally proper, it is common for ratios other than 1:1 to be referred and condensed. The nuclear chromatin of RBCs is inher-
to as if they were fractions. If the nucleus takes up less than 50% ently coarser than that of myeloid precursors, as if it is made
of the area of the cell, the proportion of nucleus is lower, and the of rope rather than yarn. It becomes even coarser and more
ratio is lower (e.g., 1:5 or less than 1). If the nucleus takes up more clumped as the cell matures, developing a raspberry-like
than 50% of the area of the cell, the ratio is higher (e.g., 3:1 or 3). appearance, in which the dark staining of the chromatin is
In the red blood cell line, the proportion of nucleus shrinks as the distinct from the almost white appearance of the parachro-
cell matures and the cytoplasm increases proportionately, although matin. This chromatin/parachromatin distinction is more
the overall cell diameter grows smaller. In short, the N:C ratio dramatic than in myeloid cells. Ultimately, the nucleus
decreases. becomes quite condensed, with no parachromatin evident
at all, and the nucleus is said to be pyknotic.
4. Nucleoli disappear. Nucleoli represent areas where the ribo-
descriptions that follow are based on the use of these types somes are formed and are seen early as cells begin actively
of stains. synthesizing proteins. As RBCs mature, the nucleoli disappear,
The stage of maturation of any blood cell is determined by which precedes the ultimate cessation of protein synthesis.
careful examination of the nucleus and the cytoplasm. The 5. The cytoplasm changes from blue to gray to pink. Blueness
qualities of greatest importance in identification of RBCs are or basophilia is due to acidic components that attract
the nuclear chromatin pattern (texture, density, homogeneity), the basic stain, such as methylene blue. The degree of
nuclear diameter, nucleus/cytoplasm (N:C) ratio (Box 8-1), cytoplasmic basophilia correlates with the amount of ribo-
presence or absence of nucleoli, and cytoplasmic color. somal RNA. These organelles decline over the life of the
CHAPTER 8 Erythrocyte Production and Destruction 89

A B C D
Figure 8-2 General trends affecting the morphology of red blood cells during the developmental process. A, Cell diameter decreases and cytoplasm changes
from blue to salmon pink. B, Nuclear diameter decreases and color changes from purplish red to dark blue. C, Nuclear chromatin becomes coarser, clumped,
and condensed. D, Composite of changes during developmental process. (Modified from Diggs LW, Sturm D, Bell A: The morphology of human blood cells,
ed 5, Abbott Park, Ill, 1985, Abbott Laboratories.)

developing RBC, and the blueness fades. Pinkness or eosino Maturation Sequence
philia is due to more basic components that attract the acid Table 8-2 lists the stages of RBC development in order and
stain eosin. Eosinophilia of erythrocyte cytoplasm correlates provides a convenient comparison. The listing makes it appear
with the accumulation of hemoglobin as the cell matures. that these stages are clearly distinct and easily identifiable.
Thus the cell starts out being active in protein production Similar to the maturing of children into adults, however, the
on the ribosomes that make the cytoplasm quite basophilic, process of cell maturation is a gradual process, with changes
transitions through a period in which the red of hemoglo- occurring in a generally predictable sequence but with some
bin begins to mix with that blue, and ultimately ends with variation for each individual. The identification of a given cells
a thoroughly pink-red color when the ribosomes are gone stage depends on the preponderance of characteristics, although
and only hemoglobin remains. the cell may not possess all the features of the archetypal
90 PART II Hematopoiesis

TABLE 8-2 Normoblastic Series: Summary of Stage Morphology


Nucleus-to-Cytoplasm % in Bone Bone Marrow
Cell or Stage Diameter Ratio Nucleoli Marrow Transit Time
Pronormoblast 12-20m 8:1 1-2 1% 24hr
Basophilic normoblast 10-15m 6:1 0-1 1-4% 24hr
Polychromatic normoblast 10-12m 4:1 0 10-20% 30hr
Orthochromic normoblast 8-10m 1:2 0 5-10% 48hr
Shift (stress) reticulocyte (polychromatic erythrocyte) 8-10m No nucleus 0 1% 48-72hr*
Polychromatic erythrocyte 8.0-8.5m No nucleus 0 1% 24-48hr*

*Transit time in peripheral blood.

50 hr 30 hr 50 hr 40 hr
Pro- Basophilic Poly- Ortho- Reticulo-
normoblast normoblast chromatic chromic cyte
normoblast normoblast
12
Average
cell 10
A diameter
(m) 8

6
RNA
Rate of content
B RNA
synthesis

Rate of
DNA
C DNA content
synthesis

34 Total protein
Hemoglobin concentration/cell
concentration 24
D (pg) 14
4 Acidophilia

Bone marrow
Figure 8-3 Timeline of cellular diameter, RNA and DNA synthesis, and hemoglobin concentration during erythropoiesis. (Modified from Granick S, Levere
RD: Heme synthesis in erythroid cells. In Moore CV, Brown EB, editors: Progress in hematology, New York, 1964, Grune & Stratton.)

descriptions that follow. Essential features of each stage are in may show small tufts of irregular cytoplasm along the periph-
italics in the following descriptions. The cellular functions ery of the membrane.
described subsequently also are summarized in Figure 8-3.
Division. The pronormoblast undergoes mitosis and gives
Pronormoblast (Rubriblast) rise to two daughter pronormoblasts. More than one division
Figure 8-4 shows the pronormoblast. is possible before maturation into basophilic normoblasts.

Nucleus. The nucleus takes up much of the cell (high N:C Location. The pronormoblast is typically present only in
ratio). The nucleus is round to oval, containing one or two the bone marrow.
nucleoli. The chromatin is open and contains few, if any, fine
clumps. Cellular Activity. The pronormoblast is beginning to
accumulate the components necessary for hemoglobin produc-
Cytoplasm. The cytoplasm is quite blue because of the con- tion. The proteins and enzymes necessary for iron uptake and
centration of ribosomes. The Golgi complex may be visible protoporphyrin synthesis are produced. Globin production
next to the nucleus as a pale, unstained area. Pronormoblasts begins.3
CHAPTER 8 Erythrocyte Production and Destruction 91

A A

B
B
Figure 8-5 A, Basophilic normoblast (prorubricyte), bone marrow (Wright
Figure 8-4 A, Pronormoblast (rubriblast), bone marrow (Wright stain,
stain, 1000). B, Electron micrograph of basophilic normoblast (15,575).
1000). B, Electron micrograph of pronormoblast (15,575). (B from Carr JH,
(B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3, Philadelphia,
Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
2009, Saunders.)

Length of Time in This Stage. This stage lasts slightly Division. The basophilic normoblast undergoes mitosis,
more than 24 hours.3 giving rise to two daughter cells. More than one division is
possible before the daughter cells mature into polychromatic
Basophilic Normoblast (Prorubricyte) normoblasts.
Figure 8-5 shows the basophilic normoblast.
Location. The basophilic normoblast is typically present
Nucleus. The chromatin begins to condense, revealing only in the bone marrow.
clumps along the periphery of the nuclear membrane and a
few in the interior. As the chromatin condenses, the parachro- Cellular Activity. Detectable hemoglobin synthesis
matin areas become larger and sharper, and the N:C ratio occurs,3 but the large number of cytoplasmic organelles, includ-
decreases to about 6:1. The staining reaction is one of a deep ing ribosomes and a substantial amount of messenger ribo-
purple-red. Nucleoli may be present early in the stage but dis- nucleic acid (RNA; chiefly for hemoglobin production),
appear later. completely mask the minute amount of hemoglobin
pigmentation.
Cytoplasm. When stained, the cytoplasm may be a deeper,
richer blue than in the pronormoblasthence the name baso- Length of Time in This Stage. This stage lasts slightly
philic for this stage. more than 24 hours.3
92 PART II Hematopoiesis

murky gray-blue. The stages name refers to this combination of


multiple colors, because polychromatophilic means many color
loving.

Division. This is the last stage in which the cell is capable


of undergoing mitosis, although likely only early in the stage.
The polychromatic normoblast goes through mitosis, produc-
ing daughter cells that mature and develop into orthochromic
normoblasts.

Location. The polychromatic normoblast is typically


present only in the bone marrow.

Cellular Activity. Hemoglobin synthesis is increasing and


the accumulation begins to be visible in the color of the cyto-
plasm. Cellular organelles are still present, particularly ribo-
somes, which contribute a blue aspect to the cytoplasm. The
progressive condensation of the nucleus and disappearance of
nucleoli are evidence of progressive decline in transcription of
A deoxyribonucleic acid (DNA).

Length of Time in This Stage. This stage lasts approxi-


mately 24 hours.3

Orthochromic Normoblast (Metarubricyte)


Figure 8-7 shows the orthochromic normoblast.

Nucleus. The nucleus is completely condensed (i.e., pyknotic)


or nearly so. As a result, the N:C ratio is quite low.

Cytoplasm. The pink-orange color of the cytoplasm reflects


nearly complete hemoglobin production. The residual ribo-
somes react with the basic component of the stain and contrib-
ute a slightly bluish hue to the cell, but that fades toward the
B
end of the stage as the organelles are degraded. The prefix ortho
Figure 8-6 A, Polychromatic normoblast (rubricyte), bone marrow means the same and refers to the fact that the cells color is
(Wright stain, 1000). B, Electron micrograph of polychromatic normoblast the same color as the eosin stain, which is red.
(15,575). (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
Philadelphia, 2009, Saunders.) Division. The orthochromic normoblast is not capable of
division due to the condensation of the chromatin.

Polychromatic (Polychromatophilic) Location. The orthochromic normoblast is typically


Normoblast (Rubricyte) present only in the bone marrow.
Figure 8-6 shows the polychromatic normoblast.
Cellular Activity. Hemoglobin production continues on
Nucleus. The chromatin pattern varies during this stage the remaining ribosomes using messenger RNA produced
of development, showing some openness early in the stage earlier. Late in this stage, the nucleus is ejected from the cell.
but becoming condensed by the end. The condensation of The cell develops pseudopod-like projections into which the
chromatin reduces the diameter of the nucleus considerably, nucleus squeezes. Ultimately, the projection separates from
so that the N:C ratio is about 4:1. Notably, no nucleoli are the cell, with the nucleus enveloped by cell membrane.4 The
present. extruded nucleus is then engulfed by splenic macrophages.
Often, small fragments of nucleus are left behind if the projec-
Cytoplasm. This is the first stage in which the redness tion is pinched off before the entire nucleus is enveloped.
associated with stained hemoglobin can be seen. The stained These fragments are called Howell-Jolly bodies when seen in
color reflects the accumulation of hemoglobin pigmentation peripheral blood cells (see Table 18-3) and are typically
over time and concurrent decreasing amounts of RNA. The removed from the circulating cells by the splenic pitting process
color produced is a mixture of pink and blue resulting in a when the cell enters the circulation.
CHAPTER 8 Erythrocyte Production and Destruction 93

A A

B B

Figure 8-7 A, Orthochromic normoblast (metarubricyte), bone marrow Figure 8-8 A, Polychromatic erythrocyte (shift reticulocyte), peripheral
(Wright stain, 1000). B, Electron micrograph of orthochromic normoblast blood (Wright stain, 1000). B, Scanning electron micrograph of polychro-
(20,125). (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3, matic erythrocyte (5000). (B from Carr JH, Rodak BF: Clinical hematology
Philadelphia, 2009, Saunders.) atlas, ed 3, Philadelphia, 2009, Saunders.)

Length of Time in This Stage. This stage lasts approxi- erythrocyte stage, the cell is the same color as a mature RBC,
mately 48 hours.3 salmon pink. It remains larger than a mature cell, however. The
shape of the cell is not the mature biconcave disc but is irregular
Polychromatic (Polychromatophilic) Erythrocyte in electron micrographs, like a lumpy potato (Figure 8-8, B).
Figure 8-8 shows the polychromatic erythrocyte. It requires the polishing activity of the spleen to assist the RBC
into its final discoid shape.
Nucleus. Beginning at the polychromatic erythrocyte stage,
there is no nucleus. The polychromatic erythrocyte is a good Division. Lacking a nucleus, the polychromatic erythro-
example of the statement that a cell may not have all the classic cyte cannot divide.
features described but may be staged by the preponderance
of features. In particular, when a cell loses its nucleus, regardless Location. The polychromatic erythrocyte resides in the
of cytoplasmic appearance, it is a polychromatic erythrocyte. marrow for 1 day or longer and then moves into the peripheral
blood, where it circulates about 1 day. During the first several
Cytoplasm. The cytoplasm can be compared easily with days after exiting the marrow, the polychromatic erythrocyte is
that of the orthochromic normoblast in that the predominant retained in the spleen for pitting and polishing by splenic mac-
color is that of hemoglobin. By the end of the polychromatic rophages, which results in the biconcave discoid mature RBC.5
94 PART II Hematopoiesis

BOX 8-2 Cellular Basophilia: Diffuse and Punctate


The reticulum of a polychromatic erythrocyte (reticulocyte) is not
seen using Wright stain. The residual RNA imparts the bluish tinge
to the cytoplasm seen in Figure 8-8, A. Based on the Wright-stained
appearance, the reticulocyte is called a polychromatic erythrocyte
because it lacks a nucleus and is no longer an erythroblast but has
a bluish tinge. When polychromatic erythrocytes are prominent on a
peripheral blood film, the examiner uses the comment polychromasia
or polychromatophilia. Wright-stained polychromatic erythrocytes are
also called diffusely basophilic erythrocytes for their regular bluish
tinge. This term distinguishes polychromatic erythrocytes from red
blood cells with punctate basophilia, in which the blue appears in
distinct dots throughout the cytoplasm. More commonly known as
basophilic stippling (see Table 18-3), punctate basophilia is associ-
ated with some anemias. Similar to the basophilia of polychromatic
erythrocytes, punctate basophilia is due to residual ribosomal RNA,
but the RNA is degenerate and stains deeply with Wright stain.

Cellular Activity. The polychromatic erythrocyte com-


pletes production of hemoglobin from residual messenger Figure 8-9 Reticulocyte, peripheral blood (new methylene blue stain,
1000).
RNA using the remaining ribosomes. The cytoplasmic protein
production machinery is simultaneously being dismantled.
Endoribonuclease, in particular, digests the ribosomes. The
acidic components that attract the basophilic stain decline over corresponds to the concavity. The pallor is about one third the
this stage to the point that the polychromatophilia is not diameter of the cell.
readily evident in the polychromatic erythrocytes on a normal
peripheral blood smear stained with Wright stain. A small Division. The erythrocyte cannot divide.
amount of residual ribosomal RNA is present, however, and
can be visualized with a vital stain such as new methylene blue, Location and Length of Time in This Stage. Mature
so called because the cells are stained while alive in suspension RBCs remain active in the circulation for approximately 120
(i.e., vital), before the smear is made (Box 8-2). The residual days.9 Aging leads to their removal by the spleen as described
ribosomes appear as a mesh of small blue strands, a reticulum, subsequently.
or, when more fully digested, merely blue dots (Figure 8-9).
When so stained, the polychromatic erythrocyte is renamed the Cellular Activity. The mature erythrocyte delivers oxygen
reticulocyte. to tissues, releases it, and returns to the lung to be reoxygen-
A second functional change in polychromatic erythrocytes ated. The dynamics of this process are discussed in detail in
is the reduced production of receptors for the adhesive mole- Chapter 10. The interior of the erythrocyte contains mostly
cules that hold developing RBCs in the marrow (see details hemoglobin, the oxygen-carrying component, and a small pro-
later).6-8 As these receptors decline, the cells are freed to leave portion of water. It has a surface-to-volume ratio and shape
the marrow. that enable optimal gas exchange to occur. If the cell were to
be spherical, it would have hemoglobin at the center of the cell
Length of Time in This Stage. The cell typically remains that would be relatively distant from the membrane and would
a polychromatic erythrocyte for about 2 days,3 with the first not be readily oxygenated and deoxygenated. With the bicon-
day spent in the marrow and the second spent in the peripheral cave shape, even hemoglobin molecules that are toward the
blood, although possibly sequestered in the spleen. center of the cell are not distant from the membrane and are
able to exchange oxygen.
Erythrocyte The cells main function of oxygen delivery throughout the
Figure 8-10 shows the erythrocyte. body requires a membrane that is flexible and deformable. The
interaction of various membrane components described in
Nucleus. No nucleus is present in mature RBCs. Chapter 9 creates these properties. RBCs must squeeze through
small spaces such as the basement membrane of the marrow
Cytoplasm. The mature circulating erythrocyte is a bicon- venous sinus. Similarly, when a cell enters the red pulp of the
cave disc measuring 7 to 8m in diameter with a thickness of spleen, it must squeeze between epithelial cells to move into
about 1.5 to 2.5m. On a stained blood smear, it appears as the venous outflow. Flexibility is crucial for RBCs to enter and
a salmon pink- or red-staining cell with a central pale area that subsequently remain in the circulation.
CHAPTER 8 Erythrocyte Production and Destruction 95

A B
Figure 8-10 A, Mature erythrocytes, peripheral blood (Wright stain, 1000). B, Scanning electron micrograph of mature erythrocytes.

The primary oxygen-sensing system of the body is located


ERYTHROKINETICS
in peritubular interstitial cells of the kidney.10,11 Hypoxia, too
Erythrokinetics is the term describing the dynamics of RBC pro- little tissue oxygen, is detected by the peritubular cells, which
duction and destruction. To understand erythrokinetics, it is produce erythropoietin (EPO), the major stimulatory cytokine
helpful to appreciate the concept of the erythron. Erythron is the for RBCs. Under normal circumstances, the amount of EPO
name given to the collection of all stages of erythrocytes produced is fairly consistent, maintaining a level of RBC pro-
throughout the body: the developing precursors in the bone duction that is sufficient to replace the approximately 1% of
marrow and the circulating erythrocytes in the peripheral RBCs that normally die each day (see section on erythrocyte
blood and the vascular spaces within specific organs such as destruction). When there is hemorrhage, increased RBC
the spleen. When the term erythron is used, it conveys the destruction, or other factors that diminish the oxygen-carrying
concept of a unified functional tissue. The erythron is distin- capacity of the blood (Box 8-3), the production of EPO can be
guished from the RBC mass. The erythron is the entirety of increased.
erythroid cells in the body, whereas the RBC mass refers only The mechanism by which hypoxia increases EPO produc-
to the cells in circulation. This discussion of erythrokinetics tion in peritubular cells is mainly transcriptional regulation.
begins by looking at the erythrocytes in the bone marrow and There is a hypoxia-sensitive region of the 3 regulatory portion
the factors that affect their numbers, their progressive develop- of the EPO gene that promotes gene transcription.12 A hypoxia-
ment, and their ultimate release into the blood. inducible factor, a transcription factor, is assembled in the
cytoplasm when oxygen tension in the cells is decreased.13 It
Hypoxiathe Stimulus to Red Blood migrates to the nucleus and interacts with the 3 enhancer for
Cell Production the EPO gene and upregulates the gene expression. With
As mentioned previously, the role of RBCs is to carry oxygen. hypoxia, more messenger RNA molecules are produced, which
To regulate the production of RBCs for that purpose, the body results in more EPO production.
requires a mechanism for sensing whether there is enough
oxygen being carried to the tissues. If not, RBC production and Erythropoietin
the functional efficiency of existing cells must be enhanced. Structure. EPO is a thermostable, nondialyzable, glyco-
Thus a second feature of the oxygen-sensing system must be a protein hormone with a molecular weight of 34kD.14 It
mechanism for influencing the production of RBCs. consists of a carbohydrate unit that reacts specifically with
96 PART II Hematopoiesis

gene activities in the RBC nucleus.18 The resulting nuclear


BOX 8-3 Hypoxia and Red Blood Cell Production activities have two major effects: allowing early release of
Teleologically speaking, the location of the bodys hypoxia sensor in reticulocytes from the bone marrow and preventing apoptotic
the kidney is practical,1 because the kidney receives approximately cell death. In addition, EPO can reduce the time needed for
20% of the cardiac output2 with little loss of oxygen. The location cells to mature in the bone marrow. These processes are
provides early detection when oxygen levels decline. Making the described in detail later. The essence is that EPO puts more
hypoxia sensor the cell that is able to stimulate red blood cell (RBC) RBCs into the circulation at a faster rate than occurs without
production also is practical, because regardless of the cause of its stimulation.
hypoxia, having more RBCs should help to overcome it. The hypoxia Early release of reticulocytes. EPO promotes early
might result from decreased RBC numbers, as with hemorrhage. release of developing erythroid precursors from the marrow.
Decreased RBC number is only one cause of hypoxia, however. Part of that results from EPO-induced changes in the adventi-
Another cause is the failure of each RBC to carry as much oxygen tial cell layer of the marrow/sinus membrane.19 Merely increas-
as it should. This can occur because the hemoglobin (Hb) is defective ing the width of the spaces through which cells might move is
or because there is not enough Hb in each cell. The hypoxia may be insufficient for cells to leave the marrow, however. RBCs are
unrelated to the RBCs in any way; poor lung function resulting in held in the marrow because they possess membrane receptors
diminished oxygenation of existing RBCs is an example. for adhesive molecules. EPO downregulates the production of
The kidneys hypoxia sensor cannot know why there is hypoxia, the receptors on the RBC surface so that cells can exit the
but it does not matter. Even when there are plenty of RBCs compared marrow earlier than they normally would.6-8 The result is the
with normal reference values, if there is still hypoxia, stimulation of presence in the circulation of reticulocytes that are still very
RBC production is warranted, because the numbers present are not basophilic, because they have not spent as much time in the
meeting the oxygen need. An elevation of RBC numbers above the marrow as they normally would. These may be called shift
normal reference values, erythrocytosis, is seen in conditions such reticulocytes because they have been shifted from the bone
as lung disease and cardiac disease in which the blood is not being marrow early (see Figure 8-8, A). Even nucleated RBCs (i.e.,
well oxygenated. Newborns have higher numbers of RBCs because normoblasts) can be released early in cases of extreme anemia
the fetal Hb in their cells does not unload oxygen to the tissues when the demand for RBCs in the peripheral circulation is
readily, and so newborns are slightly hypoxic compared with adults. great. Releasing cells from the marrow early is a quick fix, so
To compensate, they make more RBCs. to speak; it is limited in effectiveness because the available
1. Donnelly S: Why is erythropoietin made in the kidney? The kidney func- precursors in the marrow are depleted within several days and
tions as a critmeter to regulate the hematocrit, Adv Exp Med Biol still may not be enough to meet the need in the peripheral
543:73-87, 2003.
2. Stewart P: Physiology of the kidney, Update in Anaesthesia (9):1-3, 1998. blood for more cells. A more sustained response is required in
Available at: http://www.nda.ox.ac.uk/wfsa/html/u09/u09_016.htm. times of increased need for RBCs in the circulation.
Accessed May 23, 2010. Inhibition of apoptosis. A second, and probably more
important, mechanism by which EPO increases the number of
circulating RBCs is by increasing the number of cells that will
RBC receptors and a terminal sialic acid unit, which is necessary be able to mature into circulating erythrocytes. It does this by
for biologic activity in vivo.15 On desialation, EPO activity decreasing apoptosis, the programmed death of RBC progeni-
ceases.16 tors.20,21 To understand this process, an overview of apoptosis
in general is helpful.
Action. EPO is a true hormone, being produced at one Apoptosis: programmed cell death. As noted previously, it
location (the kidney) and acting at a distant location (the bone takes about 18 days to produce an RBC from stimulation of
marrow). It is a growth factor (or cytokine) that initiates an the earliest erythroid progenitor to release from the bone
intracellular message to the developing RBCs; this process is marrow. In times of increased need for RBCs, such as when
called signal transduction. EPO must bind to its receptor on the there is loss from the circulation during hemorrhage, this time
surface of cells to initiate the signal or message. Relatively few lag would be a significant problem. One way to prepare for
receptors require binding to initiate intracellular signaling.17 such a need would be to maintain a store of mature RBCs
The EPO-responsive cells vary in their sensitivity to EPO.14 somewhere in the body for emergencies. RBCs cannot be stored
Some are able to respond to low levels of EPO, whereas others in the body for this sort of eventuality, however, because they
require higher levels. In healthy circumstances when RBC pro- have a limited life span. Instead of storing cells for emergen-
duction needs to proceed at a modest but regular rate, the cells cies, the body produces cells more rapidly when needed. It
requiring only low levels of EPO respond. If EPO levels rise does so by overproduction of RBC progenitors at all times. If
secondary to hypoxia, however, a larger population of EPO- they are not needed, which is normal, the extra progenitors
sensitive cells are able to respond. are allowed to die. If they are needed, however, they have
The binding of EPO, the ligand, to its receptor on erythro- about an 8- to 10-day head start in the production process. It
cyte progenitors initiates a cascade of intracellular events is like a fast food restaurant preparing extra hamburger patties
that ultimately leads to more RBCs entering the circulation for the lunch rush just to be sure they have plenty. If the
in a given time. EPOs effects are mediated by intracellular hamburgers are not sold, they are tossed in the garbage, but if
Janus tyrosine kinase signal transducers that in turn affect they are needed, just the condiments and buns must be added
CHAPTER 8 Erythrocyte Production and Destruction 97
to make them ready to sell, which significantly reduces the time Reduced marrow transit time. Apoptosis rescue is the
needed to meet the demand. The process of intentional wastage major way in which EPO increases RBC mass, by increasing
of cells occurs by apoptosis, and it is part of the cells genetic the number of erythroid cells that survive and mature to enter
program. the circulation. The other effect of EPO is to increase the rate
Process of apoptosis. Apoptosis is a sequential process char- at which the surviving precursors can enter the circulation. This
acterized by, among other things, the degradation of chroma- is like a military recruiter who recruits more young people to
tin into large fragments of 5 to 300 kilobases that are degraded service and then puts them through a shortened boot camp
further into smaller monomers of 200 bases; protein cluster- training so that they are on the battlefield sooner. This is
ing; and activation of transglutamase. In contrast to necrosis, accomplished by two means: increased rate of cellular pro-
in which cell injury causes swelling and lysing with release of cesses and decreased cell cycle times.
cytoplasmic contents that stimulate an inflammatory response, EPO stimulates the synthesis of RBC RNA and effectively
apoptosis is not associated with inflammation.22 increases the rate of many processes, especially hemoglobin
During the sequential process of apoptosis, the following production.29 The accumulation of hemoglobin is a factor that
morphologic changes can be seen: (1) condensation of the may control other cellular processes.30 RBCs will continue to
nucleus, causing increased basophilic staining of the chroma- cycle through division and maturation until a critical concen-
tin; (2) nucleolar disintegration; and (3) shrinkage of cell tration of hemoglobin is achieved,30 typically after three to five
volume with concomitant increase in cell density and compac- divisions. Under the influence of EPO, the messenger RNA
tion of cytoplasmic organelles while mitochondria remain increases and the critical concentration of hemoglobin is
normal.23 This is followed by a partition of cytoplasm and achieved in two, rather than three, divisions. The cell is released
nucleus into membrane-bound apoptotic bodies that contain from the marrow earlier than usual. Such cells are larger, due
varying amounts of ribosomes, organelles, and nuclear mate- to a lost mitotic division, and do not have time before entering
rial. The last stage of degradation produces nuclear DNA con- the circulation to dismantle the protein production machinery
sisting of multimers of 180 base pairs. Characteristic blebbing that gives the bluish tinge to the cytoplasm. These cells are true
of the plasma membrane is observed. The apoptotic cell con- shift reticulocytes similar to those in Figure 8-8, A, recognizable
tents remain membrane-bound and are ingested by macro- in the stained peripheral blood film as especially large, bluish
phages, which avoids an inflammatory reaction. cells lacking central pallor. They also are called stress reticulo-
Evasion of apoptosis by erythroid progenitors and precursors. cytes because they exit the marrow early during conditions of
Apoptosis of RBCs is a cellular process that depends on a signal bone marrow stress, such as certain anemias. Conversely, if
from either the inside or outside of the cell. Among the crucial hemoglobin accumulates slowly, a cell may divide more times
molecules in the messaging system are the death receptor on before achieving the critical hemoglobin concentration to shut
the earliest RBC precursors, Fas, and its ligand, FasL, which is off division (Box 8-4).
expressed by more mature RBCs.23,24
When EPO levels are low, cell production should be at a
low rate, because hypoxia is not present. The excess early ery-
throid precursors should undergo apoptosis. This occurs when
the older FasL-bearing erythroid precursors, such as polychro-
BOX 8-4 Why Are Some Red Blood Cells Small
matic normoblasts, cross-link with Fas-marked immature ery-
or Large?
The decreased volume and diameter of erythrocytes seen in iron
throid precursors, such as pronormoblasts, which are then
deficiency anemia and the thalassemias can be explained by the role
stimulated to undergo apoptosis.23 As long as the more mature
of hemoglobin (Hb) in controlling cellular processes. The cell contin-
cells with FasL are present in the marrow, erythropoiesis is
ues the process of division and maturation until a critical concentra-
subdued. If the FasL-bearing cells are depleted, as when EPO-
tion of Hb is reached.1 In those anemias in which Hb production is
stimulated early release occurs, the younger Fas-positive pre-
impaired and Hb accumulates more slowly, there is time for more
cursors are allowed to develop, which increases the overall
divisions to occur before the critical concentration is reached. More
output of RBCs from the marrow.
divisions result in smaller cells. Anemias in which this occurs include
A second mechanism for escaping apoptosis exists for RBC
iron deficiency anemia in which the iron deficit prevents production
progenitors: EPO rescue. This is the major way in which EPO is
of the heme component of Hb and thalassemias in which globin
able to increase RBC production. When EPO binds to its recep-
production is impaired. The converse also would be truewhen cells
tor, one of the effects is to stimulate antiapoptotic molecules,
undergo fewer than three divisions, as is the case under the influence
which allows the cell to survive and mature.25 The cell that has
of increased levels of erythropoietin, larger cells result. This is con-
the most EPO receptors and is most sensitive to EPO is the
sistent with what is seen as patients recover from hemorrhage and
CFU-E, although the late BFU-E and early pronormoblast have
larger cells emerge from the bone marrow. Larger cells also result
some receptors.26 Without EPO, the CFU-E would not survive.27
when DNA production is impaired, leading to fewer than normal
EPOs effect is mediated by the transcription factor GATA-
numbers of divisions. This is the case with megaloblastic anemia.
1.28 The two molecules cooperate to promote the production
of the anti-apoptotic molecule bcl-xL.25 EPO-stimulated cells 1. Stohlman F: Kinetics of erythropoiesis. In Gordon AS, editor: Regulation
of hematopoiesis, vol 1, New York, 1970, Appleton-Century-Crofts,
develop this molecule on their mitochondrial membranes and
pp 318-326.
are able to resist the FasL activation of apoptosis.
98 PART II Hematopoiesis

The process by which hemoglobin exerts this influence is pig phenomenon. Although there is some evidence that
probably as a transcription factor. The presence of hemoglobin macrophages may secrete some ferritin that normoblasts
in the nucleus has been recognized for decades,31 and its role acquire,36 developing RBCs probably obtain most iron via
as a transcription factor was hypothesized even before the transferrin (see Chapter 11), and no direct contact with mac-
concept of such influential molecules had been fully deduced.30 rophages is needed for this. Because macrophages do release
It seems that hemoglobin is transferred to the nucleus in pro- ferritin, it is logical to assume that this could account for
portion to its accumulation in the cytoplasm,31 and there it the intimate arrangement. However, a more credible explana-
influences other cellular processes. tion of the cellular arrangement has been established. Macro-
EPO also can reduce the time it takes for cells to mature in phages are now known to elaborate cytokines that are vital
the marrow by reducing individual cell cycle time, specifically to the maturation process of the RBCs.37,38 RBC precursors
the length of time that cells spend between mitoses.32 This would not survive without macrophage support via such
effect is only about a 20% reduction, however, so that the stimulation.
normal transit time in the marrow of approximately 6 days A second role for macrophages in erythropoiesis also has
from pronormoblast to erythrocyte can be shortened by only been identified. Although movement of cells through the
about 1 day by this effect. marrow cords is sluggish, developing cells would exit the
With the decreased cell cycle time and fewer mitotic divi- marrow prematurely in the outflow were it not for an anchor-
sions, the time it takes from pronormoblast to reticulocyte can ing system within the marrow that holds them there until
be shortened by about 2 days. If the reticulocyte leaves the development is complete. There are three components to the
marrow early, another day can be saved, and the typical 6-day anchoring system: (1) a stable matrix of stromal cells to which
transit time is reduced to fewer than 4 days under the influence normoblasts can attach, (2) bridging (adhesive) molecules for
of increased EPO. that attachment, and (3) receptors on the erythrocyte mem-
brane. The system is analogous to anchoring a ship, with the
Measurement of Erythropoietin. Quantitative measure- anchor corresponding to a stromal cell, the anchor cable cor-
ments of EPO are performed on plasma and other body fluids. responding to the adhesive molecules, and the cable post on
EPO can be measured by immunoassays of various types, the ship corresponding to the receptor.
including radioimmunoassay and chemiluminescence. The ref- The major cellular anchor for the RBCs is the macrophage.
erence range is 5 to 30mUnits/mL.1 Increased amounts of EPO Several systems of adhesive molecules and RBC receptors tie
are expected in the urine of most patients with anemia, with the developing RBCs to the macrophages.36 At the same time,
the exception of patients with anemia caused by renal disease. RBCs are anchored to the extracellular matrix of the bone
marrow, chiefly by fibronectin.7
Other Stimuli to Erythropoiesis When it comes time for the RBCs to leave the marrow, they
In addition to tissue hypoxia, other factors influence RBC cease production of the receptors for the adhesive molecules.7
production to a modest extent. It is well documented that Without the receptor, the cells are free to move from the
testosterone directly stimulates erythropoiesis, which partially marrow into the venous sinus. Fibronectin appears to play an
explains the higher hemoglobin concentration in men than in important role in the directional migration of red blood cells
women.33 Also, pituitary34 and thyroid35 hormones have been toward the sinus.8 Entering the venous sinus requires the RBC
shown to affect the production of EPO and so have indirect to traverse the barrier created by the adventitial cells on the
effects on erythropoiesis. cord side, the basement membrane, and the epithelial cells
lining the sinus. Egress through this barrier occurs between
adventitial cells, through holes (fenestrations) in the basement
MICROENVIRONMENT OF
membrane, and through pores in the endothelial cells19
THE BONE MARROW
(Figure 8-11).39,40
The microenvironment of the bone marrow is described in
Chapter 7, and the cytokines essential to hematopoiesis are
ERYTHROCYTE DESTRUCTION
discussed there. Here, the details pertinent to erythropoiesis
(i.e., the erythropoietic inductive microenvironment) are All cells experience the deterioration of their enzymes over
emphasized, including the locale and arrangement of erythroid time due to natural catabolism. Most cells are able to replenish
cells and the anchoring molecules involved. needed enzymes and continue their cellular processes. As a
Hematopoiesis occurs in marrow cords, essentially a loose nonnucleated cell, however, the mature erythrocyte is unable
arrangement of cells in a dilated sinus area between the to generate new proteins, such as enzymes, so as its cellular
arterioles that feed the bone and the venous sinus that returns functions decline, the cell ultimately approaches death. The
blood to efferent veins. Erythropoiesis typically occurs in average RBC has sufficient enzyme function to live 120 days.
what are called erythroid islands (see Figures 7-5 and 7-6). These Because RBCs lack mitochondria, they rely on glycolysis for
are macrophages surrounded by erythroid precursors in production of adenosine triphosphate (ATP). The loss of gly-
various stages of development. It was previously believed that colytic enzymes is central to this process of cellular aging,
these macrophages provided iron directly to the normoblasts called senescence, which results in phagocytosis by macro-
for the synthesis of hemoglobin. This was termed the suckling phages. This is the major way in which RBCs die normally.
CHAPTER 8 Erythrocyte Production and Destruction 99

Figure 8-12 Macrophage ingesting a sphered erythrocyte. (From Bessis


M: Corpuscles, atlas of RBC shapes, New York, 1974, Springer-Verlag.)

of an alley. There, it is readily ingested by macrophages that


patrol along the sinusoidal lining (Figure 8-12).
The mechanisms that signal macrophages to ingest senes-
cent cells are currently under investigation. It is highly likely
that there is no single mechanism, but rather that macrophages
recognize several signals. Whether a single signal is sufficient
to elicit ingestion or whether a cell must express more than one
Figure 8-11 Egress of a red blood cell through a pore in an endothelial
cell of the bone marrow venous sinus. Arrowheads indicate the endothelial signal is uncertain. Those mechanisms receiving the greatest
cell junctions. (From DeBruyn PPH: Structural substrates of bone marrow amount of research interest include binding of autologous
function, Semin Hematol 18:182, 1981.) immunoglobulin G (IgG) to band 3 clusters,41,42 exposure of
phosphatidylserine on the exterior (plasma side) of the mem-
brane,43 and inability to maintain cation balance.44 The mac-
Macrophage-Mediated Hemolysis rophage is able to recognize these three and other characteristics
(Extravascular Hemolysis) of senescent cells that differentiate them from younger cells
At any given time, a substantial volume of blood is in the and thus targets the older cells for ingestion and lysis.
spleen, which generates an environment that is inherently When an RBC lyses within a macrophage, the major com-
stressful on cells. Movement through the red pulp is sluggish. ponents are catabolized. The iron is removed from the heme.
The available glucose in the surrounding plasma is depleted It can be stored in the macrophage as ferritin until transported
quickly as the cell flow stagnates, so glycolysis slows. The pH out. The globin of hemoglobin is degraded and returned to the
is low, which promotes iron oxidation. Maintaining reduced metabolic amino acid pool. The protoporphyrin component
iron is an energy-dependent process, so factors that promote of heme is degraded through several intermediaries to biliru-
iron oxidation cause the RBC to expend more energy and speed bin, which is released into the plasma and ultimately excreted
the catabolism of enzymes. by the liver in bile. The details of bilirubin metabolism are
In this hostile environment, aged RBCs succumb to the discussed in Chapter 22.
various stresses. Their deteriorating glycolytic processes lead to
reduced ATP production, which is complicated further by Mechanical Hemolysis
diminished amounts of available glucose. The membrane (Intravascular Hemolysis)
systems that rely on ATP begin to fail. Among these are enzymes Although most natural RBC deaths occur in the spleen, a small
that maintain the location and reduction of phospholipids of portion of RBCs rupture, normally intravascularly (within the
the membrane. Lack of ATP leads to oxidation of the mem- lumen of blood vessels). An RBCs trip around the vascular
brane lipids and proteins. Other ATP-dependent enzymes are system can be traumatic, with turbulence occurring in the
responsible for maintaining the high level of intracellular chambers of the heart or at points of bifurcation of vessels.
potassium while pumping sodium out of the cells. As this Small breaks in blood vessels and resulting clots can trap and
system fails, intracellular sodium increases and potassium rupture (i.e., lyse) cells. A few cells lyse in the blood vessels
decreases. The effect is that the selective permeability of the from purely mechanical or traumatic causes; this is called intra-
membrane is lost and water enters the cell. The discoid shape vascular hemolysis.
is lost and the cell becomes a sphere. When the membrane of the RBC has been breached, regard-
RBCs must remain highly flexible to exit the spleen by less of where the cell is located when it happens, the cell contents
squeezing through the so-called splenic sieve formed by the enter the surrounding plasma. Although mechanical lysis is a
endothelial cells lining the venous sinuses. Spherical RBCs are relatively small contributor to RBC demise under normal circum-
rigid and are not able to squeeze through the narrow spaces; stances, the body still has a system of plasma proteins, including
they become trapped against the endothelial cells and base- haptoglobin and hemopexin, to salvage the released hemo
ment membrane that form the sieve. In this situation, the RBC globin so that its iron is not lost. Hemolysis and the functions
is like a fleeing thief who is trapped against a fence at the end of haptoglobin and hemopexin are discussed in Chapter 22.
100 PART II Hematopoiesis

SUMMARY
RBCs develop from committed erythroid progenitor cells in the EPO rescues cells from apoptosis by stimulating the production
bone marrow, the BFU-E and CFU-E. of antiapoptotic molecules that counteract the effects of Fas
The morphologically identifiable precursors of mature RBCs, in and FasL.
order from youngest to oldest, are the pronormoblast, basophilic Survival of RBC precursors in the bone marrow depends on cyto-
normoblast, polychromatic normoblast, orthochromic normoblast, kines and adhesive molecules, such as fibronectin, which are
and reticulocyte. elaborated by macrophages. RBCs are found in erythroid islands,
As erythroid precursors age, the nucleus becomes condensed and where erythroblasts at various stages of maturation surround a
ultimately is ejected from the cell, which produces the reticulocyte macrophage.
stage. The cytoplasm changes color from blue, reflecting numer- As RBC precursors mature, they lose fibronectin receptors and can
ous ribosomes, to pink as hemoglobin accumulates and the ribo- leave the bone marrow. Egress occurs between adventitial cells
somes are degraded. Each stage can be identified by the extent but through pores in the endothelial cells of the venous sinus.
of these nuclear and cytoplasmic changes. Aged RBCs, or senescent cells, cannot regenerate catabolized
It takes approximately 18 days for the BFU-E to mature to an RBC, enzymes because they lack a nucleus. The semipermeable mem-
of which about 6 days are spent as identifiable precursors in the brane becomes more permeable to water, so the cell swells and
bone marrow. The mature erythrocyte has a life span of 120 days becomes spherocytic and rigid. It becomes trapped in the splenic
in the circulation. sieve.
Hypoxia of peripheral blood is detected by the peritubular cells of Extravascular or macrophage-mediated hemolysis accounts for
the kidney, which then produce EPO. most normal RBC death. The signals to macrophages that initiate
EPO, the primary hormone that stimulates the production of eryth- RBC ingestion may include binding of autologous IgG, membrane
rocytes, is able to (1) rescue the CFU-E from apoptosis, (2) shorten lipid changes, and cation balance changes.
the time between mitoses of precursors, (3) release reticulocytes Intravascular hemolysis results when mechanical factors breach
from the marrow early, and (4) reduce the number of mitoses of the cell membrane while the cell is in the peripheral circulation.
precursors. This pathway accounts for a minor component of normal destruc-
Apoptosis is the mechanism by which excessive production of tion of RBCs.
cells is controlled. Fas, the death receptor, is expressed by young
normoblasts, and FasL, the ligand, is expressed by older normo- Now that you have completed this chapter, go back and
blasts. As long as older cells mature slowly in the marrow, they read again the case study at the beginning and respond
induce the death of younger cells. to the questions presented.

R E V I E W Q UESTIONS
1. Which of the following is an erythrocyte progenitor? 4. Which of the following is not related to the effects of
a. Pronormoblast erythropoietin?
b. Reticulocyte a. The number of divisions of a normoblast
c. CFU-E b. The formation of pores in sinusoidal endothelial cells
d. Orthochromic normoblast for marrow egress
c. The time between mitoses of normoblasts
2. Which of the following is the most mature normoblast? d. The production of antiapoptotic molecules by
a. Orthochromic normoblast erythroid progenitors
b. Basophilic normoblast
c. Pronormoblast 5. Hypoxia stimulates RBC production by:
d. Polychromatic normoblast a. Inducing more pluripotent stem cells into the
erythroid lineage
3. What erythroid precursor can be described as follows: b. Stimulating EPO production by the kidney
The cell is of medium size compared with other normo- c. Increasing the number of RBC mitoses
blasts, with an N:C ratio of nearly 1:1. The nuclear chro- d. Stimulating the production of fibronectin by
matin is condensed and chunky throughout the nucleus. macrophages of the bone marrow
No nucleoli are seen. The cytoplasm is a muddy, blue-pink
color. 6. In the bone marrow, RBC precursors are located:
a. Reticulocyte a. In the center of the hematopoietic cords
b. Pronormoblast b. Adjacent to megakaryocytes along the adventitial cell
c. Orthochromic normoblast lining
d. Polychromatic normoblast c. Surrounding fat cells in apoptotic islands
d. Surrounding macrophages in erythroid islands
CHAPTER 8 Erythrocyte Production and Destruction 101

7. Which of the following determines the timing of egress of 10. Extravascular hemolysis occurs when:
RBCs from the bone marrow? a. RBCs are mechanically ruptured.
a. Maturing normoblasts slowly lose receptors for b. RBCs extravasate from the blood vessels into the
adhesive molecules that bind them to stromal cells. tissues.
b. Stromal cells decrease production of adhesive c. Splenic macrophages ingest senescent cells.
molecules over time as RBCs mature. d. Erythrocytes are trapped in blood clots outside the
c. Endothelial cells of the venous sinus form pores at blood vessels.
specified intervals of time, allowing egress of free
cells. 11. A pronormoblast belongs to the RBC mass of the body,
d. Periodic apoptosis of pronormoblasts in the marrow but not to the erythron.
cords occurs. a. True
b. False
8. What single feature of normal RBCs is most responsible
for limiting their life span? 12. A cell has an N:C ratio of 4:1. Which of the following
a. Loss of mitochondria statements would describe it?
b. Increased flexibility of the cell membrane a. The bulk of the cell is composed of cytoplasm.
c. Reduction of hemoglobin iron b. The bulk of the cell is composed of nucleus.
d. Loss of the nucleus c. The proportions of cytoplasm and nucleus are
roughly equal.
9. Intravascular hemolysis is the result of trauma to RBCs
while in the circulation.
a. True
b. False

REFERENCES
1. Dessypris EN, Sawyer ST: Erythropoiesis. In Greer JP, Foerster 13. Wang GL, Semenza GL: Characterization of hypoxia induc-
J, Rodgers GM, et al, editors: Wintrobes clinical hematology, ible factor 1 and regulation of DNA binding by hypoxia.
ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott J Biol Chem 268:21513-21518, 1993.
Williams & Wilkins, pp 106-125. 14. Kaushansky K: Hematopoietic growth factors and receptors.
2. Shumacher HR, Erslev AJ: Bone marrow kinetics. In Szirmani In Stamatoyannopoulos G, Majerus PW, Perlmutter RM,
E, editor: Nuclear hematology, New York, 1956, Academic et al, editors: The molecular basis of blood diseases, ed 3,
Press, pp 89-132. Philadelphia, 2001, Saunders, pp 25-79.
3. Granick S, Levere RD: Heme synthesis in erythroid cells. In 15. Dordal MS, Wang FF, Goldwasser E: The role of carbohydrate
Moore CV, Brown EB, editors: Progress in hematology, vol 4, in erythropoietin action. Endocrinology 116:2293-2299, 1985.
New York, 1964, Grune & Stratton, pp 1-47. 16. Goldwasser E: On the mechanism of erythropoietin-induced
4. Skutelsky E, Danon D: An electron microscopic study of differentiation: XIII. The role of sialic acid in erythropoietin
nuclear elimination from the late erythroblast. J Cell Biol action. J Biol Chem 249:4202-4206, 1974.
33:625-635, 1967. 17. Broudy VC, Lin N, Brice M, et al: Erythropoietin receptor
5. Song SH, Groom AC: Sequestration and possible maturation characteristics on primary human erythroid cells. Blood
of reticulocytes in the normal spleen. Can J Physiol Pharmacol 77:2583-2590, 1991.
50:400-406, 1972. 18. Parganas E, Wang D, Stravopodis D, et al: Jak2 is essential for
6. Patel VP, Lodish HF: The fibronectin receptor on mammalian signaling through a variety of cytokine receptors. Cell 93:385-
erythroid precursor cells: characterization and developmental 395, 1998.
regulation. J Cell Biol 102:449-456, 1986. 19. Chamberlain JK, Leblond PF, Weed RI: Reduction of
7. Vuillet-Gaugler MH, Breton-Gorius J, Vainchenker W, et al: adventitial cell cover: an early direct effect of erythropoietin
Loss of attachment to fibronectin with terminal human ery- on bone marrow ultrastructure. Blood Cells 1:655-674,
throid differentiation. Blood 75:865-873, 1990. 1975.
8. Goltry KL, Patel VP: Specific domains of fibronectin mediate 20. Sieff CA, Emerson SG, Mufson A, et al: Dependence of highly
adhesion and migration of early murine erythroid progeni- enriched human bone marrow progenitors on hemopoietic
tors. Blood 90:138-147, 1997. growth factors and their response to recombinant erythro
9. Ashby W: The determination of the length of life of transfused poietin. J Clin Invest 77:74-81, 1986.
blood corpuscles in man. J Exp Med 29:268-282, 1919. 21. Eaves CJ, Eaves AC: Erythropoietin (Ep) dose-response curves
10. Jacobson LO, Goldwasser E, Fried W, et al: Role of the kidney for three classes of erythroid progenitors in normal human
in erythropoiesis. Nature 179:633-634, 1957. marrow and in patients with polycythemia vera. Blood
11. Lacombe C, DaSilva JL, Bruneval P, et al: Peritubular cells are 52:1196-1210, 1978.
the site of erythropoietin synthesis in the murine hypoxic 22. Allen PD, Bustin SA, Newland AC: The role of apoptosis
kidney. J Clin Invest 81:620-623, 1988. (programmed cell death) in haemopoiesis and the immune
12. Semenza GL, Nejfelt MK, Chi SM, et al: Hypoxia-inducible system. Blood Rev 7:63-73, 1993.
nuclear factors bind to the enhancer element located 3 to the 23. DeMaria R, Testa U, Luchetti L, et al: Apoptotic role of Fas/
human erythropoietin gene. Proc Natl Acad Sci U S A 88:5680- Fas ligand system in the regulation of erythropoiesis. Blood
5684, 1991. 93:796-803, 1999.
102 PART II Hematopoiesis

24. Zamai L, Burattini S, Luchetti F, et al: In vitro apoptotic cell 34. Golde DW, Bersch N, Li CH: Growth hormone: species-
death during erythroid differentiation. Apoptosis 9:235-246, specific stimulation of in vitro erythropoiesis. Science
2004. 196:1112-1113, 1977.
25. Dolznig H, Habermann B, Stangl K, et al: Apoptosis protec- 35. Popovic WJ, Brown JE, Adamson JW: The influence of thyroid
tion by the Epo target Bcl-X(L) allows factor-independent hormones on in vitro erythropoiesis: mediation by a receptor
differentiation of primary erythroblasts. Curr Biol 12:1076- with beta adrenergic properties. J Clin Invest 60:908-913,
1085, 2002. 1977.
26. Sawada K, Krantz SB, Dai CH, et al: Purification of human 36. Chasis JA, Mohandas N: Erythroblastic islands: niches for
blood burst-forming unitserythroid and demonstration of erythropoiesis. Blood 112:470-478, 2008.
the evolution of erythropoietin receptors. J Cell Physiol 37. Sadahira Y, Mori M: Role of the macrophage in erythropoi-
142:219-230, 1990. esis. Pathol Int 49:841-848, 1999.
27. Koury MJ, Bondurant MC: Maintenance by erythropoietin of 38. Obinata M, Yanai N: Cellular and molecular regulation of an
viability and maturation of murine erythroid precursor cells. erythropoietic inductive microenvironment (EIM). Cell Struct
J Cell Biol 137:65-74, 1988. Funct 24:171-179, 1999.
28. Gregory T, Yu C, Ma A, et al: GAGATA-1 and erythropoietin 39. Chamberlain JK, Lichtman MA: Marrow cell egress: specificity
cooperate to promote erythroid cell survival by regulating of the site of penetration into the sinus. Blood 52:959-968,
bcl-xL expression. Blood 94:87-96, 1999. 1978.
29. Gross M, Goldwasser E: On the mechanism of erythropoietin- 40. DeBruyn PPH: Structural substrates of bone marrow func-
induced differentiation: V. Characterization of the ribonu- tion. Semin Hematol 18:179-193, 1981.
cleic acid formed as a result of erythropoietin action. Biochem 41. Kay MM, Bosman GJ, Johnson GJ, et al: Band 3 polymers and
8:1795-1805, 1969. aggregates and hemoglobin precipitates in red cell aging.
30. Stohlman F, Lucarelli G, Howard D, et al: Regulation of eryth- Blood Cells 14:275-295, 1988.
ropoiesis: XVI. Cytokinetic patterns in disorders of erythro- 42. Turrini F, Arese P, Yuan J, et al: Clustering of integral mem-
poiesis. Medicine 43:651-660, 1964. brane proteins of the human erythrocyte membrane stimu-
31. Tooze J, Davies HG: The occurrence and possible significance lates autologous IgG binding, complement deposition and
of hemoglobin in the chromosomal regions of mature eryth- phagocytosis. J Biol Chem 266:23611-23617, 1991.
rocyte nuclei of the newt Triturus cristatus cristatus. J Cell Biol 43. Boas FE, Forman L, Beutler E: Phosphatidylserine
16:501-511, 1963. exposure and red cell viability in red cell aging and in
32. Hanna IR, Tarbutt RO, Lamerton LF: Shortening of the hemolytic anemia. Proc Nat Acad Sci U S A 95:3077-3081,
cell-cycle time of erythroid precursors in response to anaemia. 1998.
Br J Haematol 16:381-387, 1969. 44. Bookchin RM, Etzion Z, Sorette M, et al: Identification and
33. Jacobson W, Siegman RL, Diamond LK: Effect of testosterone characterization of a newly recognized population of high-
on the uptake of tritiated thymidine in bone marrow of Na+, low-K+, low-density sickle and normal red cells. Proc Natl
children. Ann N Y Acad Sci 149:389-405, 1968. Acad Sci U S A 97:8045-8050, 2000.
Energy Metabolism and Membrane
Physiology of the Erythrocyte
9
Kathryn Doig and George A. Fritsma*

OUTLINE OBJECTIVES
Energy Production After completion of this chapter, the reader will be able to:
Anaerobic Glycolysis
Glycolysis Diversion 1. List red blood cell (RBC) processes that require 9. Discuss the function of glycolipids in the RBC
Pathways (Shunts) energy. membrane.
Hexose Monophosphate 2. Discuss the Embden-Meyerhof anaerobic glycolytic 10. Explain the concept of lipid exchange between the
Pathway pathway (EMP) in the erythrocyte, with attention RBC membrane and the plasma, including factors
Methemoglobin to adenosine triphosphate generation and that affect the exchange.
Reductase Pathway consumption. 11. Define, locate, and provide examples of transmem-
Rapaport-Luebering 3. Name three diversion pathways from the EMP within branous versus skeletal membrane proteins of RBCs.
Pathway RBCs and state the function or purpose of each. 12. Discuss the general structure of spectrin and its
RBC Membrane 4. Describe the role of 2,3-bisphosphoglycerate in function and arrangement in the membrane.
RBC Deformability
erythrocyte metabolism and the sacrifice made to 13. Discuss how ankyrin, protein 4.1, actin, adducin,
RBC Membrane Lipids
produce it. tropomodulin, dematin, and band 3 interact with
RBC Membrane Proteins
Osmotic Balance and 5. Name the components related to the hexose- - and -spectrin and the lipid bilayer.
Permeability monophosphate pathway that act to detoxify 14. Describe the functions of transmembranous proteins
peroxide and the type of chemical process that as they affect RBC function.
accomplishes the detoxification. 15. Cite the relative concentrations of sodium and
6. Describe the methemoglobin reductase pathway that potassium inside RBCs and name the structure that
diverts from glycolysis. maintains those concentrations.
7. Explain the importance of semipermeability of bio- 16. Name conditions that develop from abnormalities of
logic membranes. RBC membrane constituents.
8. Discuss the arrangement of lipids in the RBC
membrane.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

Cyanosis is blue skin coloration, most obvious in whites, that he treated them with vitamin C, each turned a healthy pink.
occurs when the blood does not deliver enough oxygen to Neither man was determined to have either heart or lung
the tissues. It is a common sign of heart or lung disease, in disease.
which the blood fails to become oxygenated and/or is distri 1. What does it mean to say that vitamin C is a reducing
buted improperly throughout the body. In the 1940s, Dr. agent?
James Deeny, an Irish physician, was experimenting with the 2. What must be happening if vitamin C was able to cure the
use of vitamin C (ascorbic acid), a potent reducing agent, as cyanosis?
a treatment for heart disease.1 To his disappointment, it was 3. What is the significance of finding this condition in
ineffective for nearly all patients. However, he discovered two brothers?
brothers with the distinction of being truly blue men. When

*The authors extend appreciation to Dr. John Griep, whose coverage of energy metabolism in the prior edition provided the foundation for that
content in this chapter.

103
104 PART II Hematopoiesis

T
he red blood cell (RBC, erythrocyte) transports oxygen water flow in, and the RBC swells and is destroyed. This process
from the lungs to tissues and organs and helps to trans is accelerated in hereditary nonspherocytic anemias in which
port carbon dioxide to the lungs to be exhaled. The the cells begin their life in the circulation with diminished
mammalian RBC has evolved to do this quite efficiently but enzyme concentrations.
has sacrificed greatly for this purpose by losing its nucleus.2,3 Plasma glucose enters the RBC through a facilitated mem
The resulting RBC shape facilitates oxygen acquisition and brane transport system.5 Anaerobic glycolysis, the Embden-
release. However, the cell is left without the ability to synthe Meyerhof pathway (EMP; Figure 9-1), requires glucose to
size replacement proteins for vital pathways of energy produc generate ATP, a high-energy phosphate source that is the
tion and membrane integrity, among others. As a result, its life greatest reservoir of energy in the RBC. Energy reserves like
span is limited to about 120 days because these processes glycogen are not available in RBCs, so the RBCs rely mostly on
inevitably fail. Mechanisms have evolved to maximize oxygen external glucose for glycolysis-generated ATP. Via the EMP,
exchange during the life span of the cell and to counter the glucose is catabolized to pyruvate, consuming two molecules
natural chemical processes that oxidize heme iron, which of ATP per molecule of glucose and maximally generating four
would leave it nonfunctional for oxygen transport. This chapter molecules of ATP per molecule of glucose, for a net gain of two
examines the energy production pathways of the RBC, dis molecules of ATP.
cusses mechanisms for maintaining reduced heme iron and The sequential list of biochemical intermediates involved in
facilitating oxygen delivery, and explores the features of the glucose catabolism, with corresponding enzymes, is given in
RBC membrane that contribute to effective oxygen delivery Figure 9-1. Tables 9-1 through 9-3 organize this information
for RBCs. into three phases for better comprehension.
The first phase of glucose catabolism involves glucose phos-
phorylation, isomerization, and diphosphorylation to yield fructose
ENERGY PRODUCTION
1,6-bisphosphate (F-1,6-P). F-1,6-P serves as the substrate for
ANAEROBIC GLYCOLYSIS
At the time the RBC ejects its nucleus, the cellular organelles
are also packaged and ejected from the cell. Without mitochon TABLE 9-1 Glucose Catabolism: First Phase
dria, the RBC is left to rely on glycolysis for energy production.
Substrates Enzyme Products
Fortunately, oxygen exchange and oxygen binding to heme
iron do not require energy.4 Other RBC metabolic processes do Glucose, ATP Hexokinase G6P, ADP
require energy, however, and they are listed in Box 9-1. Loss G6P Glucose phosphate isomerase F6P
of energy leads to RBC death. In fact, a hereditary deficiency of F6P, ATP Phosphofructokinase F-1,6-P; ADP
nearly every enzyme involved in glycolysis has been identified. F-1,6-P Fructodiphosphate adolase DHAP, G3P
A common result of these deficiencies is shortened RBC sur
ADP, Adenosine diphosphate; ATP, adenosine triphosphate; DHAP, dihydroxyacetone
vival, known as hereditary nonspherocytic hemolytic anemia. phosphate; F-1,6-P, fructose 1,6-bisphosphate; F6P, fructose 6-phosphate; G3P,
Transmembranous cation gradient concentration proteins, glucose 3-phosphate; G6P, glucose 6-phosphate.
called cation pumps, provide a particular example of how energy
failure, even in otherwise normal cells, leads to cell death. The
pumps maintain high concentrations of intracellular potas TABLE 9-2 Glucose Catabolism: Second Phase
sium (K+), low concentrations of sodium (Na+), and very low Substrates Enzyme Product
concentrations of calcium (Ca2+) in contrast to extracellular
concentrations of low K+ and high levels of Na+ and Ca2+. The G3P Glyceraldehyde-3-phosphate 1,3-BPG
pumps consume approximately 15% of RBC adenosine tri dehydrogenase
phosphate (ATP) production as they prevent adverse accumu 1,3-BPG, ADP Phosphoglycerate kinase 3-PG, ATP
lations of intracellular sodium and calcium. As energy-producing 1,3-BPG Bisphosphoglyceromutase 2,3-BPG
enzymes degrade over time, the pumps fail, Na+, Ca2+, and 2,3-BPG Bisphosphoglycerate phosphatase 3-PG

1,3-BPG, 1,3 Bisphosphoglycerate; 2,3-BPG, 2,3 bisphosphoglycerate; 3-PG,


3-phosphoglycerate; ADP, adenosine diphosphate; ATP, adenosine triphosphate; G3P,
glucose 3-phosphate.
BOX 9-1 Erythrocyte Metabolic Processes Requiring
Energy
Maintenance of intracellular cationic electrochemical gradients TABLE 9-3 Glucose Catabolism: Third Phase
Maintenance of membrane phospholipid
Maintenance of skeletal protein plasticity Substrates Enzyme Product
Maintenance of functional ferrous hemoglobin 3-PG Monophosphoglyceromutase 2-PG
Protection of cell proteins from oxidative denaturation 2-PG Phosphopyruvate hydratase (enolase) PEP
Initiation and maintenance of glycolysis PEP, ADP Pyruvate kinase Pyruvate, ATP
Synthesis of glutathione
Mediation of nucleotide salvage reactions 2-PG, 2-Phosphoglycerate; 3-PG, 3-phosphoglycerate; ADP, adenosine diphosphate;
ATP, adenosine triphosphate; PEP, phosphoenolpyruvate.
CHAPTER 9 Energy Metabolism and Membrane Physiology of the Erythrocyte 105
Embden-Meyerhof Pathway
(Anaerobic Pathway of Glucose Metabolism)
H2O2 H 2O Hexose Monophosphate Pathway

Glutathione peroxidase

GSH GSSG

Glucose Glutathione reductase


ATP
Hexokinase (1 ATP)
ADP NADP NADPH

Glucose 6-phosphate 6-phospho-gluconate


Glucose-6-phosphate dehydrogenase
Glucose phosphate
isomerase

Fructose 6-bisphosphate Pentose phosphate


ATP
Phosphofructokinase (1 ATP)
ADP
Fructose 1,6-bisphosphate
aldolase
Dihydroxyacetone Glyceraldehyde
phosphate 3-phosphate
Triose phosphate isomerase
Methemoglobin
Glyceraldehyde NAD reductase +H Methemoglobin
3-phosphate
dehydrogenase NADH Methemoglobin Hemoglobin
reductase

1,3-Bisphosphoglycerate Methemoglobin
Bisphosphoglycerate Reductase Pathway
mutase
ADP
(+2 ATP)
Phosphoglycerate ATP
Rapaport-Luebering
Pathway kinase

2,3-Bisphosphoglycerate
(2,3-BPG) 3-Phosphoglycerate
Bisphosphoglycerate
phosphatase

Phosphoglyceromutase

2-Phosphoglycerate

Enolase

Phosphoenolpyruvate

ADP
Pyruvate (+2 ATP)
kinase ATP

Pyruvate

NADH

NAD

Lactate
Figure 9-1 Glucose metabolism in the erythrocyte. ADP, adenosine diphosphate; ATP, adenosine triphosphate; G6PD, glucose-6-phosphate dehydrogenase;
NAD, nicotinamide adenine dinucleotide; NADH, nicotinamide adenine dinucleotide (reduced form); NADP, nicotinamide adenine dinucleotide phosphate
(oxidized form); NADPH, nicotinamide adenine dinucleotide phosphate (reduced form).
106 PART II Hematopoiesis

aldolase cleavage for the final product of phase 1 glycolysis: TABLE 9-4 Glucose Catabolism: Hexose Monophosphate
glyceraldehyde 3-phosphate (G3P; see Figure 9-1 and Table Pathway
9-1). Hexokinase and phosphofructokinase are rate-limiting in
Substrates Enzyme Product
steady-state anaerobic glycolysis, even though hexokinase has
the lowest activity of all the glycolytic enzymes. G6P Glucose-6-phosphate dehydrogenase 6-PG
The second phase of glucose catabolism converts G3P to 6-PG Phosphogluconolactonase R5P
3-phosphoglycerate (3-PG). The substrates, enzymes, and Phosphogluconate dehydrogenase
products for this phase of glycolytic metabolism are summa
rized in Table 9-2. During the first reaction step, G3P is phos 6-PG, 6-Phosphogluconate; G6P, glucose 6-phosphate; R5P, ribulose 5-phosphate.
phorylated with a high-energy phosphate and oxidized to
1,3-bisphosphoglycerate (1,3-BPG) through the action of containing cysteine. The designation GSH highlights the sulfur
glyceraldehyde-3-phosphate dehydrogenase (G3PD). 1,3-BPG moiety. Once glutathione is reduced, it is available to reduce
is dephosphorylated by phosphoglycerate kinase, which gener peroxide to water and oxygen via glutathione peroxidase.
ates ATP and 3-PG. During steady-state glycolysis, 5% to 10% of G6P is diverted
The third phase of anaerobic glucose catabolism converts to the HMP. After oxidative challenge, the activity of the HMP
3-PG to pyruvate with the generation of ATP. Substrates, may increase twentyfold to thirtyfold.6 The HMP catabolizes
enzymes, and products are listed in Table 9-3. The product 3-PG G6P to ribulose 5-phosphate and carbon dioxide by oxidizing
is isomerized by phosphoglyceromutase to 2-phosphoglycerate G6P at carbon 1. The substrates, enzymes, and products of the
(2-PG). Enolase then converts 2-PG to phosphoenolpyruvate HMP are listed in Table 9-4.
(PEP). Pyruvate kinase (PK) splits off the phosphates, forming G6PD provides the only means of generating NADPH
ATP and pyruvate. PK activity is allosterically modulated by for glutathione reduction, and in its absence erythrocytes
increased concentrations of F-1,6-P, which enhances the affinity are particularly vulnerable to oxidative damage.7 Normal
of PK for PEP.5 Thus when the F-1,6-P is plentiful, increased G6PD activity is able to detoxify oxidative compounds and
activity of PK favors pyruvate production. Pyruvic acid may safeguard hemoglobin, sulfhydryl-containing enzymes, and
diffuse from the erythrocyte or may become a substrate for membrane thiols, allowing normally functioning RBCs to carry
lactate dehydrogenase with regeneration of the oxidixed form enormous quantities of oxygen safely. However, G6PD defi
of nicotinamide adenine dinucleotide (NAD+). The ratio of ciency is the most common human RBC enzyme deficiency
NAD+ to the reduced form (NADH) may modify the activity of worldwide resulting in hereditary nonspherocytic anemia (see
this enzyme. Chapter 23).

Methemoglobin Reductase Pathway


GLYCOLYSIS DIVERSION
Heme iron is constantly exposed to oxygen, an oxidizing
PATHWAYS (SHUNTS)
agent.8 In addition, the accumulation of peroxide oxidizes
Without organelles, RBCs take advantage of diversions or heme iron from the ferrous to the ferric state, forming methe
shunts from the glycolytic pathway to generate products that moglobin. Although the HMP is able to prevent some hemo
maximize oxygen delivery from several vantage points. The globin oxidation by reducing peroxide, it is not able to reduce
three diversions are the hexose monophosphate pathway (HMP) methemoglobin once it forms. NADPH is able to do so, but
or aerobic glycolysis, the methemoglobin reductase pathway, and only slowly. The reduction of methemoglobin by NADPH is
the Rapoport-Luebering pathway. far more efficient in the presence of methemoglobin reductase,
more properly called cytochrome b5 reductase (cytob5r). Using H+
Hexose Monophosphate Pathway from NADH formed when G3P is converted to 1,3-BPG,
Aerobic or oxidative glycolysis occurs through a diversion of soluble cytochrome b5 reductase acts as an intermediate elec
glucose catabolism into the HMP, also known as the pentose tron carrier, swiftly returning the oxidized iron to its ferrous,
phosphate shunt (see Figure 9-1). The HMP detoxifies accumu oxygen-carrying state. Cytochrome b5 reductase accounts for
lated peroxide, an agent that oxidizes heme iron, proteins, and more than 65% of the methemoglobin-reducing capacity
lipids, especially those containing thiol groups. Peroxide arises within the RBC.8
spontaneously from the reduction of oxygen in the cells
aqueous environment.5 By detoxifying peroxide, the HMP Rapoport-Luebering Pathway
extends the functional life span of the RBC. A third metabolic shunt generates 2,3-bisphosphoglycerate
The HMP diverts glucose 6-phosphate (G6P) to pentose (2,3-BPG; also called 2,3-diphosphoglycerate or 2,3-DPG).
phosphate by the action of glucose-6-phosphate dehydrogenase 1,3-BPG is diverted by bisphosphoglycerate mutase to form
(G6PD). In the process, oxidized nicotinamide adenine dinu 2,3-BPG. 2,3-BPG regulates oxygen delivery to tissues by essen
cleotide phosphate (NADP+) is converted to the reduced form tially competing with oxygen for hemoglobin. When 2,3-BPG
(NADPH). NADPH is then available to reduce the oxidized binds, oxygen is released, which enhances delivery of oxygen
form of glutathione (GSSG) to its reduced form (GSH) using to the tissues.
glutathione reductase. As the thio indicates, glutathione is a 2,3-BPG forms 3-PG by the action of bisphosphoglycerate
sulfur-containing compound and is, in fact, a tripeptide phosphatase. This diversion of 1,3-BPG to form 2,3-BPG
CHAPTER 9 Energy Metabolism and Membrane Physiology of the Erythrocyte 107

immediately sacrifices the production of two ATP molecules membranes surfaces, oriented toward both the aqueous
via glycolysis. However, there is the loss of another two plasma and the cytoplasm, respectively.13 Their hydrophobic
molecules of ATP at the level of PK, because fewer molecules nonpolar acyl tails arrange themselves to form a central layer
of PEP are formed. Because two ATP molecules were used to dynamically sequestered from the aqueous plasma and cyto
generate 1,3-BPG and production of 2,3-BPG eliminates the plasm. The membrane maintains extreme differences in
production of four molecules, the cell is put in ATP deficit by osmotic pressure, cation concentrations, and gas concentra
this diversion. The cell maintains a delicate balance between tions between the plasma external to the cell and the internal
ATP generation to support the energy requirements of cell cytoplasm.14 Phospholipids reseal rapidly when the membrane
metabolism and the need to maintain the appropriate is torn.
oxygenation/deoxygenation status of hemoglobin. Acidic pH Cholesterol, esterified and largely hydrophobic, resides paral
and low concentrations of 3-PG and 2-PG inhibit the activity lel to the acyl tails of the phospholipids, equally distributed
of bisphosphoglyceromutase, thus inhibiting the shunt and between the outer and inner layers, and evenly dispersed
retaining 1,3-BPG in the EMP. These conditions and decreased within each layer, approximately one molecule per phospho
ATP activate bisphosphoglycerate phosphatase, which returns lipid molecule. Cholesterols hydroxyl group, the only hydro
2,3-BPG to the mainstream of glycolysis. In summary, these philic portion of the molecule, anchors within the polar head
conditions favor generation of ATP by causing the conversion groups, while the rest of the molecule intercalates among and
of more 1,3-BPG directly to 3-PG and also returning some 2,3 parallel to the acyl tails. Cholesterol confers tensile strength to
BPG to 3-PG for ATP generation downstream by PK. the lipid bilayer.15
The ratio of cholesterol to phospholipids remains relatively
constant and balances the need for deformability and strength.
RBC MEMBRANE
Membrane enzymes maintain the cholesterol concentration by
RBC Deformability regularly exchanging membrane and plasma cholesterol. Defi
RBCs are biconcave and average 90fL in volume.9 Their average ciencies in these enzymes are associated with membrane
surface area is 140m2, a 40% excess of surface area compared abnormalities such as acanthocytosis as the membrane loses
with a 90-fL sphere. This excess surface-to-volume ratio enables tensile strength. Conversely, as cholesterol concentration rises,
RBCs to stretch undamaged up to 2.5 times their resting diam the membrane gains strength but loses elasticity.
eter as they pass through narrow capillaries and through splenic The phospholipids are asymmetrically distributed. Phosphati-
pores 2m in diameter, a property called RBC deformability.10 dylcholine and sphingomyelin predominate in the outer layer;
The RBC plasma membrane, which is 5m thick, is 100 times phosphatidylserine (PS) and phosphatidylethanolamine form most
more elastic than a comparable latex membrane, yet has tensile of the inner layer. Distribution of the phospholipids is energy
(lateral) strength greater than that of steel. The deformable RBC dependent, relying on a number of membrane-associated
membrane provides the broad surface area and close tissue enzymes, called flippases, floppases, and scramblases, for their
contact necessary to support the delivery of oxygen from lungs position.16 When phospholipid distribution is disrupted, as in
to body tissue and carbon dioxide from body tissue to lungs. sickle cell anemia and thalassemia or in RBCs that have reached
RBC deformability depends not only on RBC geometry but the end of their 120-day life span, PS, the only negatively
also on relative cytoplasmic (hemoglobin) viscosity. The normal charged phospholipid, redistributes (flips) to the outer layer.
mean cell hemoglobin concentration (MCHC) ranges from Splenic macrophages possess receptors that bind PS and
32% to 36% (see Chapter 14 and inside front cover), and as destroy senescent RBCs. Refer to Chapter 8 for a complete dis
hemoglobin concentration rises, viscosity rises.11 Hemoglobin cussion of RBC senescence theories.
concentrations above 36% compromise deformability and Membrane phospholipids and cholesterol may also redis-
shorten the RBC life span, because the more viscous cells tribute laterally so that the RBC membrane may respond to
cannot accommodate to narrow capillaries or splenic pores. As stresses and deform within 100 milliseconds of being chal
RBCs age, they lose membrane surface area while retaining lenged by the presence of a narrow passage, such as when
hemoglobin. The hemoglobin becomes more and more con arriving at a capillary. Redistribution becomes limited as the
centrated, and eventually the RBC, unable to pass through the proportion of cholesterol increases. Plasma bile salt concentra
splenic pores, is destroyed by splenic macrophages. Refer to tion also affects cholesterol exchange.17 In liver disease with
Chapter 8 for a more complete discussion of RBC senescence low bile salt concentration, membrane cholesterol concentra
theories. tion becomes reduced. As a result, the more elastic cell mem
brane shows a target cell appearance when the RBCs are
RBC Membrane Lipids spread on a slide (see Figure 18-1).
Membrane elasticity (pliancy) is the third property that contrib Glycolipids (sugar-bearing lipids) make up 5% of the exter
utes to deformability. The RBC membrane consists of approxi nal half of the RBC membrane.18 They associate in clumps or
mately 8% carbohydrates, 52% proteins, and 40% lipids.12 The rafts and support carbohydrate side chains that extend into the
lipid portion, equal parts of cholesterol and phospholipids, aqueous plasma to help form the glycocalyx. The glycocalyx is
forms a bilayer universal to all animal cells (see Figure 13-9). a layer of carbohydrates whose net negative charge prevents
Four main phospholipids form an impenetrable fluid barrier microbial attack and protects the RBC from mechanical
as their hydrophilic polar head groups are arrayed upon the damage caused by adhesion to neighboring RBCs or to the
108 PART II Hematopoiesis

endothelium. Glycolipids may bear a few copies of carbohydrate- RBC life span. Signaling receptors bind plasma ligands and
based blood group antigens, for example, antigens of the ABH trigger energy activation of submembranous G proteins that
and the Lewis blood group systems. then initiate various energy-dependent cellular activities, a
process called signal transduction.
RBC Membrane Proteins The transmembranous proteins assemble into one of two
Although cholesterol and phospholipids constitute the princi macromolecular complexes named by their respective skeletal
pal RBC membrane structure, transmembranous (integral) and anchorages, ankyrin or protein 4.1 (Figure 9-2). These com
skeletal (cytoskeletal, peripheral) proteins make up 52% of the plexes and their anchorages provide RBC membrane structural
membrane structure by mass.19 A proteomic study reveals there integrity, because the membrane relies on the cytoplasmic
are at least 300 RBC membrane proteins, including 105 trans skeletal proteins positioned immediately within (underneath)
membranous proteins. Of the purported 300 membrane pro the lipid bilayer for its ability to retain (and return to) its
teins, about 50 have been characterized and named, some with biconcave shape despite deformability. The transmembranous
a few hundred copies per cell, others with over a million copies proteins provide vertical membrane structure.
per cell.20 Transmembranous proteins support carbohydrate-defined
blood group antigens.23 For instance, band 3 (anion transport)
Transmembranous Proteins and Glut-1 (glucose transport) support the majority of ABH
The transmembranous proteins serve a number of RBC func system carbohydrate determinants by virtue of their high copy
tions, as listed in Table 9-5.21 Through glycosylation they support numbers.24 ABH system determinants are also found on several
surface carbohydrates, which join with glycolipids to make up low copy number transmembranous proteins. Certain trans
the protective glycocalyx.22 They serve as transport and adhesion membranous proteins provide peptide epitopes. For instance,
sites and signaling receptors. Any disruption in transport protein glycophorin A carries the peptide-defined M and N determi
function changes the osmotic tension of the cytoplasm, which nants, and glycophorin B carries the Ss determinants, which
leads to a rise in viscosity and loss of deformability. Any change together comprise the MNSs system.25,26
affecting adhesion proteins permits RBCs to adhere to each The Rh system employs two multipass transmembranous
other and to the vessel walls, promoting fragmentation (vesicu lipoproteins and a multipass glycoprotein that each cross
lation), reducing membrane flexibility, and shortening the the membrane 12 times.27-30 The two lipoproteins present the

TABLE 9-5 Names and Properties of Selected Transmembranous RBC Proteins


Transmembranous Molecular Copies per % of Total
Protein Band Weight (D) Cell (103) Protein Function
Aquaporin 1 Water transporter
Band 3 (anion exchanger 1) 3 90,000-102,000 1200 27% Anion transporter, supports ABH antigens
Ca2+ ATPase Ca2+ transporter
Duffy 35,000-43,000 G proteincoupled receptor, supports Duffy antigens
Glut-1 4.5 45,000-75,000 5% Glucose transporter, supports ABH antigens
Glycophorin A PAS-1 36,000 500-1000 85% of glycophorins Transports negatively charged sialic acid, supports
MN determinants
Glycophorin B PAS-4 20,000 100-300 10% of glycophorins Transports negatively charged sialic acid, supports
Ss determinants
Glycophorin C PAS-2 14,000-32,000 50-100 4% of glycophorins Transport negatively charged sialic acid, supports
Gerbich system determinants
ICAM-4 Integrin adhesion
K+-Cl cotransporter
Kell 93,000 Zn2+-binding endopeptidase, Kell antigens
Kidd 46,000-50,000 Urea transporter
Na+,K+-ATPase
Na+-K+-2Cl cotransporter
Na+-Cl cotransporter
Na+-K+ cotransporter
Rh 30,000-45,500 D and CcEe antigens
RhAG 50,000 Necessary for expression of D and CcEe antigens;
gas transporter, probably of CO2

ATPase, Adenosine triphosphatase; Duffy, Duffy blood group system protein; ICAM, intracellular adhesion molecule; Kell, Kell blood group system protein; PAS, periodic acidSchiff
dye; RBC, red blood cell; Rh, Rh blood group system protein; RhAG, Rh antigen expression protein.
CHAPTER 9 Energy Metabolism and Membrane Physiology of the Erythrocyte 109

Ankyrin complex

Rh RhAG
Band 3 CD47

4.1R complex GPA Band 3


GPA
Duffy LW
Rh
Kell
Band 3
XK
Glut 1
GPC -spectrin -spectrin

Dematin p55 Protein 4.2 Ankyrin


Adducin
Tropomodulin
4.1R
Actin protofilament
Tropomyosin Spectrinankyrin 3
interaction

Phospholipids Membrane surface

Fatty Carbohydrates
acid
Ankyrin
chains
Integral protein
Band 3
Actin

Glycophorin


Spectrin
Peripheral protein

Spectrinactin 4.1
interaction Spectrin
dimerdimer
association

Band 4.1

Lipid
bilayer
Membrane
cytoskeleton
Figure 9-2 Representation of the human red blood cell membrane. The transmembranous proteins assemble in one of two complexes defined by their
anchorage to skeletal proteins ankyrin and 4.1. Band 3 is the most abundant transmembranous protein. In the ankyrin complex band 3 and protein 4.2 anchor
to ankyrin, which is bound to the spectrin backbone. In the 4.1 complex, band 3, Rh, and other transmembranous proteins bind the complex of dematin,
adducin, actin, tropomyosin, and tropomodulin through protein 4.1. CD47, signaling receptor; Duffy, Duffy blood group system protein; GPA, glycophorin A;
GPC, glycophorin C; Kell, Kell blood group system protein; LW, Landsteiner-Weiner blood group system protein; Rh, Rh blood group system protein; RhAG,
Rh antigen expression protein; XK, X-linked Kell antigen expression protein.
110 PART II Hematopoiesis

D and CcEe epitopes, respectively, but expression of the D


and CcEe antigens requires the separately inherited glycopro
tein RhAG, which localizes near the Rh lipoproteins in the
ankyrin complex. Loss of the RhAG glycoprotein prevents
expression of both the D and CcEd antigens (Rh-null) and is
associated with morphologic RBC abnormalities. Additional 1 -spectrin
blood group antigens localize to the 4.1 complex or specialized 2 -spectrin
proteins.28-30 2.1
A few copies of phosphatidylinositol (PI), not mentioned in 2.2 Ankyrin
the RBC Membrane Lipids section, reside in the outer, plasma-
2.3
side portion of the lipid bilayer. These serve as anchors for two
surface proteins, decay-accelerating factor (DAF, or CD55) and
membrane inhibitor of reactive lysis (MIRL, or CD59).31,32 These
proteins appear to float on the surface of the membrane as they
Adducin
link to PI through a glycan core consisting of multiple sugars.
CD55 and CD59 are linked to PI by phosphatidylinositol 3 Band 3 (anion exchanger)
glycan class A (PIG-A). In paroxysmal nocturnal hemoglobin
4.1 Protein 4.1
uria (see Chapter 23), PIG-A acquires a mutation, CD55
4.2 Protein 4.2
and CD59 become deficient, and the cell is susceptible to
complement-mediated hemolysis.
Numerical namingfor instance, band 3, protein 4.1, and
4.9 Dematin, p55
protein 4.2derives from historical (preproteomics) protein 5 Actin
identification techniques that distinguished 15 membrane pro
6 G3PD
teins using sodium dodecyl sulfate-polyacrylamide gel electrophore-
sis (SDS-PAGE), as illustrated in Figure 9-3.33 Bands migrate 7 Stomatin,
through the gel, with their velocity a property of their molecu tropomyosin
A B
lar weight and net charge, and are identified using Coomassie
blue dye. The glycophorins, with abundant carbohydrate side
chains, are stained using periodic acidSchiff (PAS) dye. Globin
Band 3, protein 4.2, and RhAG, members of the ankyrin Figure 9-3 Sodium dodecyl sulfate-polyacrylamide gel electrophoresis of
complex, link their associated proteins and the bilayer mem RBC membrane proteins stained with Coomassie blue bye. Lane A illustrates
brane to the skeletal proteins through ankyrin.34-36 Likewise, numerical band naming based on migration. Lane B names and illustrates
glycophorin C, Rh, and blood group Duffy link the 4.1 complex the positions of some of the major proteins. (Adapted from Costa FF, Agre
through protein 4.1.37 The 4.1 anchorage also includes the P, Watkins PC, et al: Linkage of dominant hereditary spherocytosis to the
more recently defined proteins adducin and dematin, which link gene for the erythrocyte membrane-skeleton protein ankyrin, N Engl J Med
with band 3 and Glut-1, respectively.38 323:1046, 1990.)

Skeletal Proteins
The principal skeletal proteins are the filamentous -spectrin
and -spectrin (Table 9-6), which assemble to form an antiparal- Joining these ends are actin and protein 4.1. Actin forms short
lel heterodimer held together with a series of lateral bonds.39 filaments of 14 to 16 monomers whose length is regulated by
Antiparallel means that the carboxyl (COOH) end of one strand tropomyosin. Adducin and tropomodulin cap the ends of
associates with the amino (NH3) end of the other. The spectrins actin, and dematin appears to stabilize actin in a manner that
form a hexagonal lattice (Figure 9-4) that is immediately adja is the subject of current investigation.43
cent to the cytoplasmic membrane lipid layer and provides Spectrin dimer bonds that appear along the length of
lateral membrane stability.40 Because the skeletal proteins the molecules disassociate and reassociate (open and close)
do not penetrate the bilayer, they are also called peripheral during RBC deformation.44 Likewise, the 20 -spectrin and 16
proteins. -spectrin repeated helices unfold and refold. These flexible
The secondary structure of both - and -spectrin features interactions plus the spectrinactinprotein 4.1 junctions are
triple-helical repeats of 106 amino acids each; 20 such repeats key regulators of membrane elasticity and mechanical stability,
make up -spectrin and 16 make up -spectrin.41 Essential to and abnormalities in any of these proteins result in deformation-
the cytoskeleton are the previously mentioned ankyrin, protein induced membrane fragmentation. For instance, autosomal
4.1, adducin and dematin, and, in addition, actin, tropomyosin, dominant mutations that affect the integrity of band 3, RhAG,
and tropomodulin (see Figure 9-4).35 A single helix at the amino ankyrin, protein 4.1, or spectrin are associated with hereditary
terminus of -spectrin consistently binds a pair of helices at spherocytosis (see Chapter 23).45,46 In these cases there are too
the carboxy terminus of the -spectrin chain, forming a stable few vertical anchorages to maintain membrane stability. The
triple helix that holds together the ends of the heterodimers.42 lipid membrane peels off in small blebs called vesicles, whereas
CHAPTER 9 Energy Metabolism and Membrane Physiology of the Erythrocyte 111

TABLE 9-6 Names and Properties of Selected Skeletal RBC Proteins


Copies per % of Total
Skeletal Protein Band Molecular Weight (D) Cell (103) Protein Function
-spectrin 1 240,000-280,000 240 16% Filamentous antiparallel heterodimer,
-spectrin 2 220,000-246,000 240 14% primary cell skeleton
Adducin 2.9 80,000-103,000 60 4% Caps actin filament
Ankyrin 2.1 206,000-210,000 120 4.5% Anchors band 3 and protein 4.2
Dematin 4.9 43,000-52,000 40 1% Actin bundling protein
F-actin 5 42,000-43,000 400-500 5.5% Binds -spectrin
G3PD 6 35,000-37,000 500 3.5%
Protein 4.1 4.1 66,000-80,000 200 5% Anchors 4.1R complex
Protein 4.2 (protein kinase) 4.2 72,000-77,000 200 5% Anchors ankyrin complex
Tropomodulin 5 41,000-43,000 30 Caps actin filament
Tropomyosin 7 27,000-38,000 80 1% Regulates actin polymerization

G3PD, Glucose-3-phosphate dehydrogenase glyceraldehyde; RBC, red blood cell.

Actin
Spectrin
dimer
Actin
Band 3

Adducin

Band 4.1

Band 4.1

A Tropomyosin B
Glycophorin

Junctional
complex

Spectrin dimer
Ankyrin 100 nm
Figure 9-4 Spectrin-based cytoskeleton on the cytoplasmic side of the human red blood cell membrane. A, Junctional complex composed of actin filaments
containing 14 to 16 actin monomers, band 4.1, adducin, and tropomyosin, which probably determines the length of the actin filaments. B, Spectrin dimers
form a lattice that binds band 3 and protein 4.2 (not shown) via ankyrin and band 3, Glut-1 and Duffy (not shown), and glycophorin via protein 4.1.

the cytoplasmic volume remains intact. This generates sphero cytoplasm.49 Aquaporin 1 is a transmembranous protein that
cytes with a reduced membrane-to-cytoplasm ratio. Conversely, forms pores or channels whose surface charges create inward
hereditary elliptocytosis (ovalocytosis) arises from one of several flow of water in response to internal osmotic changes. Glucose
autosomal dominant mutations affecting spectrin dimer-to- is transported without energy expenditure by the transmem
dimer lateral bonds or the spectrinankyrinprotein 4.1 junc branous protein Glut-1.
tion.47 In hereditary elliptocytosis, the membrane fails to Adenosine triphosphatase (ATPase)dependent (energy-
rebound from deformation, and RBCs progressively elongate dependent) cation pumps (see Table 9-5) regulate the concentra
to form visible elliptocytes, which causes a mild to severe tions of Na+ and K+, maintaining intracellular-to-extracellular
hemolytic anemia.48 ratios of 1:12 and 25:1, respectively.50,51 These enzyme-based
pump mechanisms, in addition to aquaporin, maintain
Osmotic Balance and Permeability osmotic balance. Pump mechanism damage permits the influx
The RBC membrane is impermeable to Na+, K+, and Ca2+. It is of Na+, with water following osmotically. The cell swells,
permeable to water and the anions bicarbonate (HCO3) and becomes spheroid, and eventually ruptures, spilling hemoglo
chloride (Cl), which freely exchange between plasma and RBC bin. This phenomenon is called colloid osmotic hemolysis.
112 PART II Hematopoiesis

Ca2+ ATPase extrudes calcium, maintaining exceptionally Sickle cell disease provides an example of increased cation
low intracellular levels of 5 to 10mol/L. Calmodulin, permeability. When crystallized sickle hemoglobin deforms
a cytoplasmic Ca2+-binding protein, controls the function the cells, internal levels of Na+, K+, and especially Ca2+ rise,
of Ca2+ ATPase.52 which results in hemolysis.53

SUMMARY
Glucose enters the erythrocyte by facilitated membrane The membrane proteins include the transmembranous (integral)
transport. proteins. These include ion channels and various receptors.
Glycolysis in the EMP occurs aerobically and anaerobically. The proteins that localize to the interior surfaces of the membrane
The primary reservoir of energy is ATP, which is generated by are called skeletal (peripheral) proteins.
glycolysis. Membrane proteins are extracted using sodium dodecyl sulfate,
Through anaerobic glycolysis in the EMP, glucose is metabolized separated using polyacrylamide gel electrophoresis, and stained
to pyruvate, using two molecules of ATP per molecule of glucose with Coomassie blue for visualization. They are numbered from
and providing four molecules of ATP per molecule of glucose, for the point of application; lower numbers correlate to high protein
a net gain of two molecules of ATP. molecular weight and lower net charge.
By means of the Rapaport-Luebering pathway (a shunt off the PAS staining is used to localize electrophoresed proteins with high
EMP), 2,3-BPG is generated. It is necessary for the facilitation of carbohydrate composition, which are designated as PAS-1 through
oxygen delivery to the tissues. PAS-4.
The methemoglobin reductase pathway (also a bypass from the The shape and flexibility of the RBC, which are essential to its
EMP) is able to convert oxidized heme iron (methemoglobin) to its function, are related to the cytoskeleton. The cytoskeleton is
reduced state, returning the molecule to its oxygen-carrying derived from a group of peripheral proteins on the interior side of
function. the lipid membrane. The major structural proteins are - and
Aerobic glycolysis occurs in the HMP, which converts glucose into -spectrin, which are bound together and to the lipid membrane
pentose with the generation of NADPH. NADPH reduces glutathi- by ankyrin, actin, protein 4.1, adducin, tropomodulin, dematin, and
one, which then reduces peroxide to water and oxygen. Proteins, band 3.
lipids, and heme iron are then protected from oxidation by Abnormalities of the peripheral structural proteins such as spectrin
peroxide. and actin lead to abnormal RBC shapes. Hereditary spherocytosis
The RBC membrane is a typical lipid bilayer with hydrophobic and hereditary elliptocytosis are examples of diseases associated
components oriented away from the water-rich plasma and with abnormalities and deficiencies of structural proteins.
cytoplasm. The concentration of potassium inside the RBC is higher than that
The two lipid layers of the membrane differ in the composition of in the plasma, whereas the sodium concentration is lower. This
phospholipids. disequilibrium is maintained by the Na+-K+ pump in the mem-
The membrane provides a semipermeable barrier separating the brane. Failure of the pump leads to an influx of sodium, with water
plasma constituents from the cytoplasm, which allows the cell to following by osmosis, which results in cell swelling and possible
maintain necessary intracellular differences. lysis.
Membrane cholesterol is restored from the plasma by an enzyme-
enhanced process of lipid exchange. Now that you have completed this chapter, go back and
Abnormalities in membrane lipids lead to abnormal cell shapes, read again the case study at the beginning and respond
such as the target cells that develop with liver disease, which to the questions presented.
result from the changes in plasma bile salts.

R E V I E W Q UESTIONS
1. An RBC process that does not require energy is: 2. ATP is generated by the:
a. Maintenance of intracellular cationic electrochemical a. Embden-Meyerhof pathway
gradients b. Hexose monophosphate pathway
b. Oxygen transport c. Luebering-Rapaport pathway
c. Maintenance of skeletal protein plasticity d. Aerobic glycolytic pathway
d. Initiation and maintenance of glycolysis
CHAPTER 9 Energy Metabolism and Membrane Physiology of the Erythrocyte 113

3. Which of the following is true concerning 2,3-BPG? 9. The numbering system for membrane proteins gives the
a. It enhances the release of oxygen from hemoglobin. lowest number to proteins that are:
b. It provides a source of glucose for the RBC. a. Smallest
c. It is unnecessary for RBC survival. b. Most negatively charged
d. It is the least abundant of all organophosphates in c. Largest
the RBC. d. Most positively charged

4. The activity of the hexose monophosphate pathway 10. Which of the following is an example of an integral mem
increases the RBC source of: brane protein?
a. Glucose and lactic acid a. Spectrin
b. 2,3-BPG and methemoglobin b. Glycophorin A
c. NADPH and reduced glutathione c. Actin
d. ATP and other purine metabolites d. Ankyrin

5. The layer of the erythrocyte membrane that is largely 11. Which of the following statements about spectrin structure
responsible for the shape, structure, and deformability of is correct?
the cell is the: a. Spectrin forms homodimers that twist into ropelike
a. Integral protein layer strands.
b. Exterior lipid layer b. Spectrin dimers associate into tetramers that are
c. Peripheral protein layer tightly bound to each other throughout their length.
d. Interior lipid layer c. Seven spectrin tetramers join at their ends to create a
lattice.
6. The glycolipids of the RBC membrane: d. The spectrin strands are connected to the lipid bilayer
a. Provide flexibility to the membrane by ankyrin.
b. Constitute ion channels
c. Carry RBC antigens 12. All of the following are functions of band 3 except:
d. Attach the cytoskeleton to the lipid layer a. Providing a channel for anion movement through the
membrane
7. The lipids of the membrane are arranged: b. Contributing to membrane flexibility
a. In chains beneath a protein exoskeleton c. Attaching spectrin to the lipid bilayer
b. So that hydrophobic portions are facing the d. Identifying senescent RBCs
plasma
c. In a hexagonal lattice 13. Na+,K+-ATPase maintains a concentration of ions in which:
d. In two layers that are not symmetric in composition a. Potassium is in higher concentration inside the RBC
than in the plasma, and sodium is in lower
8. Lipid exchange between the RBC membrane and the concentration in the RBC than in the plasma
plasma occurs: b. Sodium and potassium are in higher concentration
a. To replace lost lipids in the membrane inside the RBC than in the plasma
b. To provide a mechanism for excretion of lipid-soluble c. Sodium is in higher concentration inside the RBC
RBC waste products than in the plasma, and potassium is in equilibrium
c. To ensure symmetry between the composition of the with the plasma
interior and exterior lipid layers d. Potassium and sodium are in lower concentration
d. To provide lipid-soluble nutrients to the RBC inside the RBC than in the plasma

REFERENCES
1. Percy MJ, Lappin TR: Recessive congenital methaemoglobi Williams hematology, ed 7, New York, 2006, McGraw-Hill,
naemia: cytochrome b5 reductase deficiency. Brit J Haem pp 603-631.
141:298-308, 2008. 5. Kondo T, Beutler E: Developmental changes in glucose trans
2. Bull B: Morphology of the erythron. In Lichtman MA, Beutler port of guinea pig erythrocytes. J Clin Invest 65:1-4, 1980.
E, Kipps TJ, et al, editors: Williams hematology, ed 7, 6. Prchal JT, Gregg JT: Cell enzymopathies. In Hoffman R, Benz
New York, 2006, McGraw-Hill, pp 369385. EJ, Shattil SJ, et al, editors: Hematology principles and practice,
3. Thelen MJ: The mature erythrocyte. In Greer JP, Foerster J, ed 3, New York, 2000, Churchill Livingstone, pp 561-576.
Rodgers GM, et al, editors: Wintrobes clinical hematology, 7. Beutler E: Disorders of red cells resulting from enzyme abnor
ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott malities. Semin Hematol 27:137167, 1990.
Williams & Wilkins, pp 126-155. 8. Beutler E: Methemoglobinemia and other causes of cyanosis.
4. Beutler E: Disorders of red cells resulting from enzyme abnor In Lichtman MA, Beutler E, Kipps TJ, et al, editors: Williams
malities. In Lichtman MA, Beutler E, Kipps TJ, et al, editors: hematology, ed 7, New York, 2006, McGraw-Hill, pp 701-708.
114 PART II Hematopoiesis

9. Mohandas N, Gallagher PG: Red cell membrane: past, 33. Fairbanks G, Steck TL, Wallach DF: Electrophoretic analysis
present, and future. Blood 112:3939-3948, 2008. of the major polypeptides of the human erythrocyte mem
10. Schmid-Schnbein H: Erythrocyte rheology and the optimi brane. Biochemistry 10:26062617, 1971.
zation of mass transport in the microcirculation. Blood Cell 34. Jennings ML: Topical review: oligometric structure and the
1:285-306, 1975. anion transport function of human erythrocyte band 3
11. Mohandas N, Chasis JA: Red blood cell deformability, mem protein. J Membr Biol 80:105117, 1984.
brane material properties, and shape: regulation by trans 35. Kay MM, Bosman GJ, Johnson GJ, et al: Band 3 polymers and
membrane, skeletal and cytosolic proteins and lipids. Semin aggregates and hemoglobin precipitates in red cell aging.
Hematol 30:171-192, 1993. Blood Cells 14:275295, 1988.
12. Cooper RA: Lipids of human red cell membrane: normal 36. Bennett V: Proteins involved in membrane-cytoskeleton
composition and variability in disease. Semin Hematol 7:296- association in human erythrocytes: spectrin, ankyrin, and
322, 1970. band 3. Methods Enzymol 96:313324, 1983.
13. Singer SJ, Nicolson GL: The fluid mosaic model of the struc 37. Anderson RA, Lovrien RE: Glycophorin is linked by band 4.1
ture of cell membranes. Science 175:720-731, 1972. protein to the human erythrocyte membrane skeleton. Nature
14. Discher DE: New insights into erythrocyte membrane orga 307:655658, 1984.
nization and microelasticity. Curr Opin Hematol 7:117122, 38. Khan AA, Hanada T, Mohseni M, et al: Dematin and adducin
2000. provide a novel link between the spectrin cytoskeleton and
15. Cooper RA: Influence of the increased membrane cholesterol human erythrocyte membrane by directly interacting with
on membrane fluidity and cell function in human red blood glucose transporter-1. J Biol Chem 283:1460014609, 2008.
cells. J Supramol Struct 8:413-430, 1978. 39. Shotton DM, Burke BE, Branton D: The molecular structure
16. Zhou Q, Zhao J, Stout JG, et al: Molecular cloning of human of human erythrocyte spectrin: biophysical and electron
plasma membrane phospholipid scramblase. A protein microscope studies. J Mol Biol 131:303329, 1979.
mediating transbilayer movement of plasma membrane 40. Liu SC, Drick LH, Palek J: Visualization of the hexagonal
phospholipids. J Biol Chem 272:18240-18244, 1997. lattice in the erythrocyte membrane skeleton. J Cell Biol
17. Cooper RA, Jandl JH: Bile salts and cholesterol in the patho 104:527536, 1987.
genesis of target cells in obstructive jaundice. J Clin Invest 41. Speicher DW, Marchesi VT: Erythrocyte spectrin is composed
47:809822, 1968. of many homologous triple helical segments. Nature
18. Alberts B, Johnson A, Lewis J, et al: Membrane structure. In 311:177180, 1984.
Alberts B, Johnson A, Lewis J, et al, editors: Molecular biology 42. Shen BW, Josephs R, Steck TL: Ultrastructure of unit frag
of the cell, ed 5, 2007, Garland Science. ments of the skeleton of the human erythrocyte membrane.
19. Steck T: The organization of proteins in the human red blood J Cell Biol 99:810821, 1984.
cell membrane. J Cell Biol 62:119, 1974. 43. An X, Guo X, Zhang X, et al: Conformational stabilities of the
20. Pasini EM, Kirkegaard M, Mortensen P, et al: In-depth analy structural repeats of erythroid spectrin and their functional
sis of the membrane and cytosolic proteome of red blood implications. J Biol Chem 281:1052710532, 2006.
cells. Blood 108:791801, 2006. 44. DeSilva TM, Peng KC, Speicher KD, et al: Analysis of human
21. Bennett V: The membrane skeleton of human erythrocytes red cell spectrin tetramer (head-to-head) assembly using
and its implications for more complex cells. Annu Rev Biochem complementary univalent peptides. Biochemistry 31:10872
54:273304, 1985. 10878, 1992.
22. Furthmayr H: Glycophorins A, B, and C. A family of sialogly 45. An X, Lecomte MC, Chasis JA, et al: Shear-response of the
coproteins, isolation and preliminary characterization of spectrin dimer-tetramer equilibrium in the red blood cell
trypsin derived peptides. In Lux SE, Marchesi VT, Fox CJ, membrane. J Biol Chem 277:3179631800, 2002.
editors: Normal and abnormal red cell membranes, New York, 46. Costa FF, Agre P, Watkins PC, et al: Linkage of dominant
1979, Alan R Liss, pp 195-211. hereditary spherocytosis to the gene for the erythrocyte
23. Reid ME, Mohandas N: Red blood cell blood group antigens: membrane-skeleton protein ankyrin. N Engl J Med 323:1046
structure and function. Semin Hematol 41:93117, 2004. 1050, 1990.
24. Daniels G: ABO, Hh, and Lewis systems. In Human blood 47. Gallagher PG, Forget BG: Hematologically important muta
groups, ed 2, Oxford, 2002, Blackwell, pp 7-98. tions: spectrin and ankyrin variants in hereditary spherocyto
25. Daniels G: MNS blood group system. In Human blood groups, sis. Blood Cells Mol Dis 24:539543, 1998.
ed 2, Oxford, 2002, Blackwell, pp 99-173. 48. Coetzer T, Zail S: Spectrin tetramer-dimer equilibrium in
26. Daniels G: Gerbich blood group system. In Human blood hereditary elliptocytosis. Blood 59:900905, 1982.
groups, ed 2, Oxford, 2002, Blackwell, pp. 426-443. 49. Brugnara C: Erythrocyte membrane transport physiology.
27. Daniels G: RH blood group system. In Human blood groups, Curr Opin Hematol 4:122127, 1997.
ed 2, Oxford, 2002, Blackwell, pp. 195-274. 50. Jorgensen PL: Mechanism of the Na+, K+ pump: protein struc
28. Daniels G: Duffy blood group system. In Human blood groups, ture and conformation of the pure (Na+ K+)-ATPase. Biochim
ed 2, Oxford, 2002, Blackwell, pp. 324-351. Biophys Acta 694:2768, 1982.
29. Daniels G: Kell and KX blood group systems. In Human blood 51. James PH, Pruschy M, Vorherr TE, et al: Primary structure of
groups, ed 2, Oxford, 2002, Blackwell, pp. 295-323. the cAMP-dependent phosphorylation site of the plasma
30. Daniels G: Kidd blood group system. In Human blood groups, membrane calcium pump. Biochemistry 28:42534258, 1989.
ed 2, Oxford, 2002, Blackwell, pp. 342-351. 52. Takakuwa Y, Mohandas N: Modulation of erythrocyte mem
31. Beutler E. Paroxysmal nocturnal hemoglobinuria. In Lichtman brane material properties by Ca2+ and calmodulin: implica
MA, Beutler E, Kipps TJ, et al, editors: Williams hematology, tions for their role in skeletal protein interaction. J Clin Invest
ed 7, New York, 2006, McGraw-Hill, pp. 469-475. 82:394400, 1988.
32. Daniels G: Cromer blood group system. In Human blood 53. Rhoda MD, Aprovo N, Beuzard Y, et al: Ca2+ permeability in
groups, ed 2, Oxford, 2002, Blackwell, pp. 444-454. deoxygenated sickle cells. Blood 75:24532458, 1990.
Hemoglobin Metabolism
Mary Coleman
10
OUTLINE OBJECTIVES
Hemoglobin Structure After completion of this chapter, the reader will be able to:
Heme Structure
Globin Structure 1. Describe the primary structure of the globin chains 13. Identify the P50 value (amount of oxygen needed to
Complete Hemoglobin found in hemoglobin. saturate 50% of hemoglobin) as it pertains to a
Molecule 2. Describe the quaternary structure of hemoglobin. normal oxygen dissociation curve.
Hemoglobin Biosynthesis 3. Describe the biosynthesis of heme and globin. 14. Differentiate T and R forms of hemoglobin.
Heme Biosynthesis 4. Differentiate steps in heme synthesis that occur in 15. Identify the source of production of 2,3-
Globin Biosynthesis the mitochondria and the cytoplasm. bisphosphoglycerate and describe its impact on
Hemoglobin Assembly 5. Identify the ontogeny of hemoglobin with emphasis hemoglobin oxygenation.
Hemoglobin Ontogeny on the hemoglobin of newborns and adults. 16. Identify the oxygen affinity of fetal hemoglobin.
Hemoglobin Production 6. Identify the three types of normal hemoglobin in 17. Compare and contrast the composition of the
Regulation
adults and their reference intervals. chemically modified hemoglobinsmethemoglobin,
Heme Regulation
7. Describe the regulatory effects of hemoglobin carboxyhemoglobin, and sulfhemoglobinand their
Globin Regulation
Hemoglobin Regulation metabolism. affinity for oxygen.
Hemoglobin Function 8. Identify the important role that hemoglobin plays in 18. Compare and contrast oxygenated, deoxygenated, and
Variant Hemoglobins maintaining body functions. oxidized hemoglobin and ferric versus ferrous iron.
Methemoglobin 9. Describe the mechanism by which hemoglobin 19. Describe how hemoglobin is routinely measured in
Sulfhemoglobin carries oxygen to the tissue. the laboratory.
Carboxyhemoglobin 10. Describe the Bohr effect. 20. Identify how different kinds of hemoglobins are iden-
Hemoglobin Measurement 11. Explain the significance of the sigmoid shape of the tified by laboratory tests.
oxygen dissociation curve. 21. Identify the gene locations of the globins that make
12. Correlate right and left shifts in the oxygen dissocia- up the hemoglobin molecule, including the number
tion curve with conditions that can cause shifts in of genes for each globin chain (for Hb A, A2, and F)
the curve. and their general arrangement on chromosomes.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

Hemoglobin and hemoglobin electrophoresis testing were 1. Were these hemoglobin results within expected reference
performed on a mother and her newborn infant, both pre intervals?
sumed to be healthy. The assays were part of a screening 2. Why were the mothers and the newborns hemoglobin
program to establish reference values. The mothers hemo results so different?
globin value was 14g/dL (140g/L), and the infants was 3. What is the difference between a hemoglobin test and the
20g/dL (200g/L). The mothers hemoglobin electrophoresis hemoglobin electrophoresis test?
results were 97% Hb A, 2% Hb A2, and 1% Hb F. The 4. Why were the mothers and newborns hemoglobin
newborns results were 88% Hb F and 12% Hb A. electrophoresis results so different?

groups and two heterogenous pairs of polypeptide chains


HEMOGLOBIN STRUCTURE
(Figure 10-1).
Hemoglobin (Hb) is the first protein whose structure was Hemoglobin is the main cytoplasmic component of eryth
described using x-ray crystallography.1-4 The hemoglobin mole rocytes (red blood cells, or RBCs). Free (non-RBC) hemoglo
cule is a conjugated globular protein consisting of four heme bin, generated from RBCs through hemolysis, has a short

115
116 PART II Hematopoiesis

1 2 2

Heme

1 2 bonds and 2 1 bonds

Heme

Heme
1 1 are located between the dimers,
1 1 bonds are in the front,
2 2 bonds are behind.

Heme
2 2

2 1
2
1 1

Figure 10-1 Hemoglobin: a tetramer of four heme molecules, each attached to a globin polypeptide chain.

CH2 TABLE 10-1 Globin Chains


=

CH CH3 Symbol Name No. of Amino Acids


H
C
Alpha 141
CH3 CH=CH2
Beta 146
N N A Gamma A 146 (position 136: alanine)
G Gamma G 146 (position 136: glycine)
HC Fe2+ CH
Delta 146
N N
Epsilon Unknown
Zeta 141
CH3 CH3
Theta Unknown
C
H
CH2 CH2
give rise to different types of polypeptide chains. Each chain is
HOOC CH2 CH2 COOH designated by a Greek letter (Table 10-1).5
Figure 10-2 Heme is protoporphyrin IX that carries a central ferrous Each globin chain is divided into eight helices and seven
ion (Fe2+). nonhelical segments (Figure 10-3). The helices, designated A
to H, contain subgroup numberings for the sequence of the
amino acids in each helix and are relatively rigid and linear.
half-life, is rapidly salvaged, and is catabolized or excreted The flexible nonhelical segments connect the helices, as
renally. The concentration of hemoglobin within RBCs is reflected by their designations: NA for the sequence between
approximately 34g/dL, and its molecular weight is 64,000 D the N-terminus and the A helix, AB between the A and B
(Daltons). Hemoglobins main function is to transport oxygen helices, and so forth with BC, CD, DE, EF, FG, GH, and finally
from the lungs to tissues. Hemoglobin also modulates vascular HC between the H helix and the C-terminus.
dilation by transporting nitric oxide (NO) and transports
carbon dioxide from the tissues to the lungs for exhalation. Complete Hemoglobin Molecule
The hemoglobin molecule can be described by its primary, sec
Heme Structure ondary, tertiary, and quaternary protein structures. The primary
Heme consists of a ring of carbon, hydrogen, and nitrogen structure refers to the amino acid sequence of the polypeptide
atoms called protoporphyrin IX with an atom of divalent ferrous chains. The secondary structure refers to chain arrangements
iron (Fe2+) attached (ferroprotoporphyrin, Figure 10-2). Each in helices and nonhelices. The tertiary structure refers to the
of the four heme groups is positioned in a pocket of the poly arrangement of the helices into a pretzel-like configuration.
peptide chain near the surface of the hemoglobin molecule. Globin chains loop to form a cleft pocket for heme. Each
Each heme molecule combines reversibly with one oxygen chain contains a heme group that is suspended between the E
molecule. Owing to its double bonds, heme renders blood red. and F helices of the polypeptide chain. The iron atom at the
center of the protoporphyrin IX ring of heme is positioned
Globin Structure between two histidine radicals, forming a proximal histidine
The four globin chains comprising each hemoglobin molecule bond within F8 and, through the linked oxygen, a close asso
consist of two identical pairs of unlike polypeptide chains, 141 ciation with the distal histidine residue in E7. The distal histi
to 146 amino acids each. Variations in amino acid sequences dine appears to swing in and out of position to permit the
CHAPTER 10 Hemoglobin Metabolism 117

C Terminus ALA PRO PRO


LYS
ASN ALA GLN ALA
HIS ALA
ALA GLY GLN
VAL
THR
TYR LYS ALA VAL VAL TYR PHE
HIS VAL
LEU 130
140 H GLU

120 LYS

G ALA
GLY
ASN CYS HIS
VAL PHE
GLU ARG LEU VAL
GLY VAL LEU HIS
100 LEU
PRO ASN LEU
PHE 110
ASP

VAL
HIS LEU LYS
VAL N Terminus ASP

ALA
F 90
HIS GLU CYS
THR SER
PHE
GLY THR LEU HIS
LEU LYS
LEU
THR LEU

GLU PRO 80 ASN F8 (Proximal histidine)


GLU LYS ASP
10 SER LEU
HIS E
ALA A GLY 70
VAL ALA ASP
THR ALA E7 (Distal histidine residue)
ALA LEU PHE
LEU TRP SER LYS
GLY VAL
GLY HIS
LYS LEU
20 LYS ALA
VAL LYS
Close spatial contact GLY
VAL ASP GLY VAL
LYS
ALA LEU LEU 60 PRO
ASN VAL GLY
LEU ASN
GLU VAL
GLU GLY ARG TYR
TRP GLY
VAL
B PRO
THR
C
D VAL MET
GLN PHE ALA
ARG ASP
PHE
40 GLU 50 THR PRO

SER PHE GLY ASP LEU SER

Figure 10-3 A -globin chain: a polypeptide with helical and nonhelical segments. (From Huisman TH, Schroder WA: New aspects of the structure,
function, and synthesis of hemoglobins, Boca Raton, Fla, 1971, CRC Press; modified from Stamatoyannopoulos G: The molecular basis of blood diseases,
ed 2, Philadelphia, 1994, Saunders.)

passage of oxygen into and out of the hemoglobin molecule. The complete hemoglobin molecule is spherical, has four
Globin chain amino acids in the cleft are hydrophobic, whereas heme groups attached to four polypeptide chains, and may
amino acids on the outside are hydrophilic, which renders the carry four molecules of oxygen. It is composed of two globin
molecule water soluble. This arrangement also helps iron chains and two non- globin chains. Each globin chain has a
remain in its divalent ferrous form regardless of whether it is heme group attached. Each heme molecule is capable of carry
oxygenated (carrying oxygen molecules) or deoxygenated (not ing one molecule of oxygen. Strong 1, non-1 and 2,non-2
carrying oxygen molecules). dimeric bonds hold the molecule in a stable form. Tetrameric
The quaternary structure of hemoglobin, also called a tetra 1non-2 and 2,non-1 bonds also contribute to the stabil
meric molecule, describes the complete hemoglobin molecule. ity of the structure (Figure 10-4).6,7
118 PART II Hematopoiesis

Heme

C
1 Heme
D F

F C
B
E
E G H 2
G
H
B D
A A

A H
F
B E
A
G
G

C
E H B Heme
C
Heme

F
D
non2 non1

Figure 10-4 Hemoglobin molecule illustrating tertiary folding and quaternary assembly. Heme is suspended between the E and F helices of the polypeptide
chain. Pink represents 1 (left) and 2 (right); yellow represents non-2 (left) and non-1 (right).

IX to make heme. Heme leaves the mitochondria and is joined


HEMOGLOBIN BIOSYNTHESIS
to the globin chains in the cytoplasm.
Heme Biosynthesis
Heme biosynthesis occurs in the mitochondria and cytoplasm
of bone marrow RBC precursors, beginning with the pronor Globin Biosynthesis
moblast (also known as proerythroblast) through the circu Six structural genes control the synthesis of the six globin
lating polychromatic (also known as polychromatophilic) chains. The and genes are on chromosome 16; the , , ,
erythrocyte (see Chapter 8). As they lose their mitochondria and genes are linked on chromosome 11. In the human
and the citric/tricarboxylic acid cycle, mature RBCs can no genome, there is one copy of each globin gene per chromatid
longer make hemoglobin. for a total of two genes per person with the exception of and
Heme biosynthesis begins with the condensation of glycine . There are two copies of the and genes per chromatid for
and succinyl coenzyme A (CoA) catalyzed by aminolevulinate syn a total of four genes per person.
thase (ALAS) to form aminolevulinic acid (ALA). ALA dehydratase The production of globin chains takes place in RBC pre
(also known as ALA dehydrase, porphobilinogen synthase) in cursors from the pronormoblast through the circulating poly
the presence of ALA catalyzes the formation of porphobilinogen. chromatic (polychromatophilic) erythrocyte, but not in the
Porphobilinogen deaminase, also known as hydroxymethylbilane mature RBC.8,9 Globin proteins arise via transcription of
synthase, in the presence of porphobilinogen catalyzes the the genetic code to messenger ribonucleic acid (mRNA) and
formation of hydroxyl methylbilane. This pathway continues translation of mRNA to the globin polypeptide chain. A slight
until, in the final step of production of heme, Fe2+ combines excess of -globin mRNA is present in pronormoblasts (pro
with protoporphyrin IX in the presence of ferrochelatase/heme erythroblasts); however, -globin mRNA is translated more
synthase to make heme (Figure 10-5).6 efficiently than -globin mRNA. This results in synthesis of
Transferrin, a plasma protein, carries iron in the ferric (Fe3+) sets of chains in approximately equal amounts. When syn
form to developing RBCs. Iron passes through the RBC mem thesized, the chains are released from the ribosomes in the
brane to the mitochondria and is united with protoporphyrin cytoplasm.7
CHAPTER 10 Hemoglobin Metabolism 119

MITOCHONDRION CYTOSOL

Glycine
ALA synthase ALA dehydratase
 ALA Porphobilinogen (PBG)
Succinyl CoA
PBG deaminase

Hydroxymethylbilane
Uroporphyrinogen
III synthase
Uroporphyrinogen III
Uroporphyrinogen
decarboxylase
Coproporphyrinogen oxidase
Protoporphyrinogen IX Coproporphyrinogen III
Protoporphyrinogen
oxidase
Protoporphyrin IX

Ferrochelatase
 Fe2
Heme

Heme Heme
 
 polypeptide Non- polypeptide
chain chain
Ribosomes Ribosomes

Non Heme

, non- dimer
Heme
Heme

Non

Heme

Non
Heme

Heme

Non 2, non-2 tetramer



Heme
Heme

Figure 10-5 Hemoglobin assembly begins with glycine and succinyl coenzyme A (CoA), which assemble in the mitochondria catalyzed by aminolevulinic
acid (ALA) synthase to form ALA. In the cytoplasm, ALA undergoes several transformations to form coproporphyrin III, which, catalyzed by coproporphyrinogen
oxidase, becomes protoporphyrinogen IX. In the mitochondria protoporphyrinogen IX is converted to protoporphyrin IX by protoporphyrinogen oxidase. Ferrous
(Fe2+) ion is added, catalyzed by ferrochetolase to form heme. In the cytoplasm, heme assembles with chain and non- chain globins to form dimers and
ultimately hemoglobin tetramers.

Hemoglobin Assembly hemoglobin in postnatal life. Hb A2 contains two and two


After their release from ribosomes, each globin chain binds to chains. The chains are inefficiently expressed; only small
a heme molecule and they pair off. An chain and a non- amounts of Hb A2 are found in the RBCs. Hb F contains two
chain combine to form heterodimers. Two heterodimers spon and two chains. In adults, Hb F is distributed unevenly
taneously combine to form tetramers. The tetrameric 12 and among RBCs; it is present in a few RBCs called F cells.7
21 bonds also contribute to the stability of the structure. This The various globin chains affect the net negative charge of
completes the hemoglobin molecule (see Figure 10-5).6,7 the hemoglobin molecule. In hemoglobin electrophoresis,
The combination of two and two chains along with the hemoglobins under the influence of an electrical field exhibit
four heme molecules forms Hb A. This is the predominant varying mobilities, which allows differentiation of hemoglobin
120 PART II Hematopoiesis

Birth
Months 1 2 3 4 5 6 7 8 2 4 6 8 10

Site of
blood cell Yolk Liver Bone marrow
production sac

Spleen

A
50

40
Globin
chain 30
synthesis
% of total 20


10

Weeks 0 10 20 30 0 10 20 30 40

Intrauterine life Birth Infant


B
Figure 10-6 Timeline of globin chain production from intrauterine life to adulthood. See also Table 10-2.

types. Various support media, buffer, and pH are employed for TABLE 10-2 Normal Hemoglobins
definitive identification (see Chapter 26).
Stage Globin Chain Hemoglobin
Intrauterine
Early embryogenesis (product of yolk 2 + 2 Gower-1
HEMOGLOBIN ONTOGENY
sac erythroblasts) 2 + 2 Gower-2
Hemoglobin composition differs with prenatal gestation time 2 + 2 Portland
and postnatal age. Hemoglobin changes reflect changes in the Begins in early embryogenesis; peaks 2 + 2 F
activation and inactivation or switching of the globin genes, during midgestation and declines
progressing from the to the gene on chromosome 16 and rapidly just before birth
from the to the , , or genes on chromosome 11. The
and genes normally appear only during the first 3 months of Birth
embryonic development. These two chains in addition to the 2 + 2 F, 60-90%
and chains are constituents of embryonic hemoglobins 2 + 2 A, 10-40%
(Figure 10-6). At birth, Hb F is the predominant hemoglobin.
Normal adult hemoglobin is predominately Hb A (22) with Adulthood
small amounts of Hb A2 (22) and Hb F (22).7 Table 10-2 2 + 2 F, 1-2%
presents adult reference intervals. 2 + 2 A2, <3.5%
A small percentage of Hb A is glycated. Glycation is 2 + 2 A, >95%
a posttranslational modification formed by nonenzymatic
binding of various sugars with globin chain amino groups.
The most glycated hemoglobin is Hb A1c, in which glucose
HEMOGLOBIN PRODUCTION REGULATION
attaches to the N-terminal valine of the chain. Normally,
about 4% to 6% of Hb A circulates in the A1c form. In Heme Regulation
uncontrolled diabetes mellitus, the amount of A1c is increased. The key rate-limiting step in heme synthesis is the initial re
Other posttranslational modifications seem to be of little action of glycine and succinyl CoA to form ALA, catalyzed by
importance.7 ALAS. Transcription of the ALAS gene is inhibited by heme,
CHAPTER 10 Hemoglobin Metabolism 121
which leads to a decrease in heme production (a negative
100
feedback mechanism). Other enzymes in the heme pathway

in
inhibited by heme are ALA dehydrase and porphobilinogen

lob

7.6
deaminase/hydroxymethylbilane synthase. An increased demand 80

og
My

pH
for heme induces an increased synthesis of ALAS.9
Ferrochelatase (also known as heme synthase) also plays a B

% O2 saturation

7.2
60
regulatory role in heme biosynthesis. A negative feedback

pH
7.4
mechanism by heme or substrate inhibition by protoporphyrin P50

pH
IX is believed to inhibit the ferrochelatase/heme synthase 40 C
A
enzyme.9

20 Hemoglobin
Globin Regulation
Globin production is regulated by the rate at which the deoxy
ribonucleic acid (DNA) is transcribed to mRNA. The amount
of the specific globins synthesized is proportional in general to 0 20 40 60 80 100
the content of their individual globin mRNAs. Heminthe Fe3+ PO2 (mm Hg)
oxidation product of hemeis important in controlling the Figure 10-7 Oxygen dissociation curve. A, Normal dissociation curve.
rate of globin synthesis in intact polychromatic erythrocytes B, Left-shifted curve with reduced P50 caused by a decrease in
and various cell-free systems, and in its absence, an inhibitor 2,3-bisphosphoglycerate (2,3-BPG), partial pressure of carbon dioxide (PCO2),
of globin synthesis accumulates. Balanced globin chain and temperature, and H+ ions (raised pH). A left-shifted curve is also seen with
heme synthesis is important, because excess components of hemoglobin variants that have increased oxygen affinity. C, Right-shifted
hemoglobinunpaired chains, protoporphyrin, and iron curve with increased P50 caused by an elevation in 2,3-BPG, PCO2, tempera-
ture, and H+ ions (lowered pH). A right-shifted curve is also seen with
reduce RBC survival. Normal mature RBCs contain only com
hemoglobin variants that have decreased oxygen affinity.
plete hemoglobin molecules; pools of free heme or globin
chains are minute.7,9
called the P50 value. The relationship is described by the
Hemoglobin Regulation oxygen dissociation curve of hemoglobin, which plots the
Hemoglobin synthesis is normally stimulated by tissue hypoxia. percent oxygen saturation of hemoglobin versus the PO2 (Figure
Hypoxia causes the kidneys to produce increased erythropoi 10-7). The curve is sigmoidal, which indicates low hemoglobin
etin, which stimulates the production of hemoglobin and affinity for oxygen at low oxygen tension and high affinity for
RBCs. Although each laboratory must establish its own refer oxygen at high oxygen tension.
ence intervals based on their instrumentation, methodology, Cooperation among hemoglobin subunits contributes to
and patient population, in general, reference intervals for the shape of the curve. Heme units do not undergo simultane
hemoglobin are as follows: ous oxygenation or deoxygenation; rather, the state of each
heme unit in comparison with the other units influences further
Men: 14 to 18g/dL (140 to 180g/L)
binding. That is, hemoglobin that is completely deoxygenated
Women: 12 to 15g/dL (120 to 150g/L)
has little affinity for oxygen, but with each oxygen molecule
Newborns: 16.5 to 21.5g/dL (165 to 215g/L)
that is bound, the avidity increases, and the hemoglobin mole
Reference intervals for infants and children vary according cule quickly becomes fully oxygenated.7 Shifts of the curve to
to age group. Individuals living at high altitudes have slightly the left or right occur if there are changes in the pH of the blood.
higher levels of hemoglobin as a compensatory mechanism to This shift in the curve as a result of pH is termed the Bohr effect.
provide more oxygen to the tissues in the oxygen-thin air. Normally, a PO2 of approximately 27mmHg results in
Tables on the inside front cover of this text provide reference 50% oxygen saturation of the hemoglobin molecule. If there
intervals for all age groups. is a shift of the curve to the left, 50% oxygen saturation of
hemoglobin occurs at a PO2 of less than 27mmHg. If there is
a shift of the curve to the right, 50% oxygen saturation of
HEMOGLOBIN FUNCTION
hemoglobin occurs at a PO2 higher than 27mmHg.
The function of hemoglobin is to readily bind oxygen mole The reference interval for arterial oxygen saturation is 96%
cules in the lung, which requires high oxygen affinity; to trans to 100%. If the oxygen dissociation curve shifts to the left, a
port oxygen; and to efficiently unload oxygen to the tissues, patient with arterial and venous PO2 concentrations in the refer
which requires low oxygen affinity. During oxygenation, each ence intervals (80 to 100mmHg arterial and 30 to 50mmHg
of the four heme iron ions in a hemoglobin molecule can venous) will have a higher percent oxygen saturation and a
reversibly bind one oxygen molecule. Approximately 1.34mL higher affinity for oxygen than a patient for whom the curve is
of oxygen is bound by each gram of hemoglobin. normal. With a shift in the curve to the right, a lower oxygen
The affinity of hemoglobin for oxygen depends on the affinity is seen. Hemoglobin absorbs less oxygen in the lungs
partial pressure of oxygen (PO2), often defined in terms of the but delivers more oxygen to the tissues than it normally would,
amount of oxygen needed to saturate 50% of hemoglobin, as in the presence of Hb S (see Chapter 26).
122 PART II Hematopoiesis

2 2
1 2

Heme 15 Heme O 2
2,3-BPG
1

e
Hem
Heme

Heme
1 1

e
Hem
O2
1

O2
Heme Heme O 2
2 2
2 1

2 1 1 2

Deoxygenated (tense, T) state Oxygenated (relaxed, R) state


Figure 10-8 Tense (T) and relaxed (R) forms of hemoglobin. The tense form incorporates one 2,3-bisphosphoglycerate (2,3-BPG) molecule and salt bridges
and is unable to transport oxygen. The relaxed form develops as 2,3-BPG is released, the salt bridges are broken, and oxygen molecules are incorporated.

The effect of 2,3-bisphosphoglycerate (2,3-BPG, formerly Myoglobin is released into the plasma when there is damage
2,3-diphosphoglycerate), on oxygen affinity is complex. The to the muscle in myocardial infarction, trauma, or severe
hemoglobin molecule is allosteric; that is, its function and muscle injury, called rhabdomyolysis. Myoglobin is normally
structure are influenced by other molecules. In the presence of excreted through the kidney, but levels may become elevated
large amounts of 2,3-BPG, the hemoglobin molecule changes in renal failure. Testing for serum myoglobin aids in detecting
from a relaxed (R) oxygenated molecule to a tense (T) deoxy myocardial infarction in patients who have no underlying
genated molecule. The T structure is stabilized by salt bridges, trauma, rhabdomyolysis, or renal failure. An elevated myoglo
which become broken as the molecule switches to the R struc bin level generates a positive result on the urine hemoglobin
ture. When the salt bridges are broken, the hemoglobin mole dipstick test that must be differentiated from a response caused
cule is able to bind to oxygen (Figure 10-8). Carbon dioxide, by hemoglobin. In contrast to myoglobinuria, hemoglobinuria
hydrogen ions, and chloride ions all decrease the affinity of is seen in intravascular hemolytic disorders.7
hemoglobin for oxygen by strengthening the salt bridges that Hb F (fetal hemoglobin, the primary hemoglobin in new
lock the molecule into its T conformation. Some abnormal borns) has a P50 of 19 to 21mmHg, which results in a left shift
hemoglobins with a high oxygen affinity and low P50 occur as of the oxygen dissociation curve and increased affinity relative
a result of amino acid substitutions that lead to loss of bonds to that of Hb A. Consequently, fetal circulation extracts oxygen
that would have stabilized the tetramer in the T conformation. from maternal blood. Hb F delivers oxygen less readily to
Without these binding sites, the hemoglobin molecule holds tissues, however. To achieve adequate tissue oxygenation, fetal
on more tightly to oxygenhence a higher oxygen affinity.7 hemoglobin concentration is high. The concentration approxi
Clinical conditions that produce a shift to the left include mates adult values by 6 months as Hb F production is reduced.
a lowered body temperature due to external causes; multiple A second crucial function of hemoglobin is the transport
transfusions of stored blood with depleted 2,3-BPG; alkalosis; of carbon dioxide. In the blood, carbon dioxide undergoes a
and the presence of methemoglobin, carboxyhemoglobin, or pair of reactions in which it combines with water to form car
some other hemoglobin variants. Conditions producing a shift bonic acid. Carbonic acid then disassociates to release H+ and
of the curve to the right include increased body temperature, bicarbonate:
increased 2,3-BPG concentrations, increased H+ concentration
CO2 + H2O H2CO3
(decreased pH), and abnormal hemoglobins with a low affinity
H2CO3 H+ + HCO3
for oxygen. Clinical conditions that produce a right shift include
a high fever, acidosis, and circumstances that produce hypoxia, The first of these reactions is facilitated by the RBC enzyme
such as high altitude, pulmonary insufficiency, congestive heart carbonic anhydrase. The H+ from the second reaction binds
failure, severe anemia, and cardiac right-to-left shunt.7 deoxygenated hemoglobin. The bicarbonate diffuses across the
The sigmoidal oxygen curve generated by normal hemoglo RBC membrane, and a portion is exchanged with plasma Cl;
bin contrasts with myoglobins hyperbolic curve (see Figure this is called the chloride shift. The plasma bicarbonate travels
10-7). Myoglobin, present in cardiac and skeletal muscle, is a to the lungs, where it is expired. About 70% to 85% of tissue
17,000-D monomeric oxygen-binding heme protein. It binds carbon dioxide is processed in this manner.10 Approximately
oxygen with greater affinity than hemoglobin. Its hyperbolic 10% to 20% of carbon dioxide binds to the N-terminal amino
curve indicates that it releases oxygen only at very low partial group of each globin chain as carbaminohemoglobin. Carb
pressures, which means it is not as effective as hemoglobin aminohemoglobin has a lower affinity for oxygen than does
in releasing oxygen to the tissues at physiologic tensions. hemoglobin in the absence of carbon dioxide.11
CHAPTER 10 Hemoglobin Metabolism 123

spectral curve does not shift when cyanide is added, a feature


VARIANT HEMOGLOBINS
that is used to distinguish it from methemoglobin.13
The variant hemoglobins methemoglobin, sulfhemoglobin,
and carboxyhemoglobin are hemoglobins whose structure has Carboxyhemoglobin
been modified by drugs or environmental chemicals. Carboxyhemoglobin results from the combination of carbon mon
oxide with heme iron. Although carbon monoxide binds hemo
Methemoglobin globin more slowly than oxygen, its affinity is 240 times that of
Methemoglobin is a form of hemoglobin that contains iron in oxygen and its release is 10,000 times slower. Carbon monoxide
the oxidized or ferric state (Fe3+). Methemoglobin is continu has been termed the silent killer because it is an odorless and
ously being formed by spontaneous oxidation, but fails to colorless gas, and victims may quickly become hypoxic.5,13
accumulate because several reducing enzyme systems restrict Some carboxyhemoglobin is produced endogenously. The
its concentration to less than 1% of total hemoglobin reference interval is 0.2% to 0.8%. Exogenous carbon monox
(see Chapter 9). ide is derived from the exhaust of automobiles and from indus
Methemoglobin is brownish to bluish and does not revert trial pollutants, such as coal, gas, charcoal burning, and tobacco
to red upon oxygen exposure. Ferric iron cannot bind oxygen, smoke. In smokers, levels may vary from 4% to 20%.5,13 Expo
but when one or more ferric ions are present the conformation sure to carbon monoxide may be coincidental, accidental, or
of the molecule changes, and the oxygen affinity of the remain intentional (suicidal). Many deaths from house fires are the
ing heme groups increases.12,13 Increased methemoglobin pro result of inhaling smoke, fumes, or carbon monoxide.16 Even
duces a shift to the left in the oxygen dissociation curve, so that when heating systems in the home are properly maintained,
oxygen is not delivered efficiently to the tissues. If methemo accidental poisoning with carbon monoxide may occur.17 Toxic
globin comprises more than 30% of total hemoglobin, patients effects, such as headaches and dizziness, are experienced at
present with hypoxia and cyanosis.5,14 levels of 10% to 15%. Levels of more than 50% may cause
Elevated levels of methemoglobin are seen when oxidants coma and convulsions. Carboxyhemoglobin may be detected
such as nitrites are present or when there is decreased activity by spectral absorption instruments at 541nm. It gives blood a
of methemoglobin reductase, a genetic deficiency. It also is cherry-red color, which is also imparted to the skin of victims.
seen in patients who inherit Hb M disease, which is caused by Hyperbaric oxygen therapy has been used for treatment.
an abnormality in the structure of the globin portion of the
hemoglobin molecule (see Chapter 26).15
HEMOGLOBIN MEASUREMENT
Methemoglobin is assayed by spectral absorption analysis
instruments such as the CO-oximeter. Methemoglobin shows The cyanmethemoglobin method is the reference method for
a peak in the range of 620 to 640nm at a pH of 7.1. Acquired hemoglobin assay.18 A lysing agent present in the cyanmethe
methemoglobinemia may be treated by removal of the moglobin reagent frees hemoglobin from RBCs. Free hemoglo
offending substance or administration of ascorbic acid or bin combines with potassium ferricyanide contained in the
methylene blue.15 cyanmethemoglobin reagent, which converts the hemoglobin
iron from the ferrous to the ferric state to form methemoglobin.
Sulfhemoglobin Methemoglobin combines with potassium cyanide to form the
Sulfhemoglobin is formed by the irreversible oxidation of hemo stable pigment cyanmethemoglobin. The cyanmethemoglobin
globin by sulfonamides, phenacetin, acetanilide, or phenazo color intensity, which is proportional to hemoglobin concen
pyridine. It is created in vitro by the addition of hydrogen tration, is measured at 540nm spectrophotometrically and
sulfide to hemoglobin and has a greenish pigment. Sulfhemo compared with a standard (see Chapter 14). The cyanmethe
globin is ineffective for oxygen transport, and patients with moglobin method is performed manually but has been adapted
elevated levels present with cyanosis. Sulfhemoglobin cannot for use in automated instruments.
be converted to normal adult hemoglobin; it persists for the Many instruments now use sodium lauryl sulfate (SLS) to
life of the cell. Treatment consists of prevention by avoidance convert hemoglobin to SLS-methemoglobin. This method does
of the offending agent. not generate toxic wastes (see Chapter 39).
Sulfhemoglobin, like methemoglobin, shows a peak at Hemoglobin electrophoresis is used to separate the different
620nm on a spectral absorption instrument. The sulfhemoglobin types of hemoglobins such as Hb A, A2, and F (see Chapter 26).

SUMMARY
The hemoglobin molecule is composed of four heme groups (pro- Hemoglobin biosynthesis is regulated by hormones, oxygen
toporphyrin IX + Fe2+) and two pairs of unlike polypeptide chains. tension in the kidneys, and enzymes in the heme synthesis
Each heme molecule combines with one polypeptide chain. pathway.
Hemoglobin, contained in RBCs, carries oxygen to the tissue 2,3-BPG produced by the glycolytic pathway facilitates the delivery
bound to the ferrous iron in heme. of oxygen from hemoglobin to the tissues.
124 PART II Hematopoiesis

The oxygenation dissociation curve of hemoglobin is sigmoid Chemically modified hemoglobins do not transport oxygen to the
owing to cooperativity among the hemoglobin subunits. tissues well, which results in cyanosis.
The Bohr effect is the influence of pH on the hemoglobin oxygen
release mechanism. Now that you have completed this chapter, go back and
The three hemoglobins found in normal adults are Hb A, Hb A2, read again the case study at the beginning and respond
and Hb F. Hb A, composed of two heterodimers, is the to the questions presented.
predominant hemoglobin of adults.
Hemoglobin ontogeny describes which hemoglobins are produced
by the body from the fetal period through birth to adulthood.

R E V I E W Q UESTIONS
1. A hemoglobin molecule is composed of: 7. What is the distribution of normal hemoglobins in
a. One heme molecule and four globin chains adults?
b. Ferrous iron, protoporphyrin IX, and a globin a. 80% to 90% Hb A, 5% to 10% Hb A2, 1% to
chain 5% Hb F
c. Protoporphyrin IX and four globin chains b. 80% to 90% Hb A2, 5% to 10% Hb A, 1% to
d. Four heme molecules and four globin chains 5% Hb F
c. >95% Hb A, <3.5% Hb A2, 1% to 2% Hb F
2. Normal adult Hb A contains which polypeptide chains? d. >90% Hb A, 5% Hb F, 1% Hb A2
a. and
b. and 8. Which of the following is a description of the structure of
c. and oxidized hemoglobin?
d. and a. Hemoglobin carrying oxygen on heme; synonymous
with oxygenated hemoglobin
3. A key rate-limiting step in heme synthesis is suppression b. Hemoglobin with iron in the ferric state
of: (methemoglobin) and not able to carry oxygen
a. Aminolevulinate synthase c. Hemoglobin with iron in the ferric state so that
b. Transferrin mRNA synthesis carbon dioxide replaces oxygen in the heme
c. Iron oxidase structure
d. Protoporphyrin IX reductase d. Hemoglobin carrying carbon monoxide; hence the
oxidized refers to the single oxygen
4. Which of the following forms of hemoglobin molecule has
the lowest affinity for oxygen? 9. In the quaternary structure of hemoglobin, the globin
a. Tense chains associate into:
b. Relaxed a. tetramers in some cells and tetramers in others
c. Arterial b. A mixture of tetramers and tetramers in each
d. Venous cell
c. dimers and dimers
5. Using the normal hemoglobin oxygenation curve in Figure d. Two dimers
10-7 for reference, predict the position of the oxygenation
curve for methemoglobin. 10. How are the globin chain genes arranged?
a. Shifted to the right of normal a. With genes and genes on the same chromosome,
b. Shifted to the left of normal including two genes and two genes
c. No change b. With genes and genes on separate chromosomes,
including two genes on one chromosome and one
6. The predominant hemoglobin found in a normal newborn gene on a different chromosome
is: c. With genes and genes on the same chromosome,
a. Gower-1 including four genes and four genes
b. Gower-2 d. With genes and genes on separate chromosomes,
c. A including four genes on one chromosome and two
d. F genes on a different chromosome
CHAPTER 10 Hemoglobin Metabolism 125

11. The nature of the interaction between 2,3-BPG and c. Binds to amino acids of the globin chain,
hemoglobin is that 2,3-BPG: contributing to a conformational change that inhibits
a. Binds to the heme moiety, blocking the binding of oxygen from binding to heme
oxygen d. Oxidizes hemoglobin iron, diminishing oxygen
b. Binds simultaneously with oxygen to ensure that it binding and promoting oxygen delivery to the tissues
stays bound until it reaches the tissues, when both
molecules are released from hemoglobin

REFERENCES
1. Perutz MF: Molecular anatomy, physiology and pathology of 10. Hsia CCW: Respiratory function of hemoglobin. N Engl J Med
hemoglobin. In Stamatoyannopoulos G, Nienhuis AW, Leder 338:239248, 1998.
P, editors: The molecular basis of blood diseases, Philadelphia, 11. Telen MJ: The mature erythrocyte. In Greer JP, Foerster J,
1987, Saunders, pp 127-173. Rodgers GM, et al, editors: Wintrobes clinical hematology,
2. Perutz MF, Rossman MG, Cullis AP, et al: Structure of ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott
hemoglobin: a three dimensional Fourier synthesis at 5.5A Williams & Wilkins, pp 126-155.
resolution obtained by x-ray analysis. Nature 185:416422, 12. Darling RC, Roughton FJW: The effect of methemoglobin on
1960. the equilibrium between oxygen and hemoglobin. Am J
3. Perutz MF, Kendrew JC, Watson HC: Structure and function Physiol 137:5668, 1946.
of hemoglobin: II. Some relations between polypeptide chain 13. Steinberg MH: Hemoglobins with altered oxygen affinity,
con?guration and amino acid sequence. J Mol Biol 13:669- unstable hemoglobins, M-hemoglobins, and dyshemo
678, 1965. globinemias. In Greer JP, Foerster J, Rodgers GM, et al,
4. Perutz MF, Muirhead H, Cox JM, et al: Three dimensional editors: Wintrobes clinical hematology, ed 12, Philadelphia,
Fourier synthesis of horse oxyhaemoglobin at 2.8 resolu 2009, Wolters Kluwer Health/Lippincott Williams & Wilkins,
tion: the atomic model. Nature 219:131139, 1968. pp 1132-1142.
5. Mazzella FM Schumacher HR: Hemoglobin. In Kaplan LA, 14. Benz EJ Jr: Hemoglobin variants associated with hemolytic
Pesce AJ, editors: Clinical chemistry theory, analysis, correlation, anemia, altered oxygen affinity, and methemoglobinemias.
ed 5, St Louis, 2010, Mosby, pp 771-789. In Hoffman R, Furie B, McGlave P, et al, editors: Hema
6. Deacon AC, Whatley SD, Elder GH: Porphyrins and disorders tology basic principles and practice, ed 5, Philadelphia,
of porphyrin metabolism. In Burtis CA, Ashwood ER, 2009, Churchill Livingstone: Available at MD Consult:
Bruns DE, editors: Tietz textbook of clinical chemistry and http://www.mdconsult.com/book/player/book.do?method=
molecular diagnostics, ed 4, Philadelphia, 2006, Saunders, display&type=aboutPage&decorator=header&eid=4-u1.0-
pp 1209-1235. B978-0-443-0671, Accessed October 2, 2009.
7. Steinberg MH, Benz EJ Jr, Adewoye HA, et al: Pathobiology 15. Beutler E: Methemoglobinemia and other causes of cyanosis.
of the human erythrocyte and its hemoglobins. In Hoffman In Lichtman MA, Beutler E, Kipps TJ, et al, editors: Williams
R, Furie B, McGlave P, et al, editors: Hematology basic hematology, ed 7, New York, 2006, McGraw-Hill Professional,
principles and practice, ed 5, Philadelphia, 2009, Churchill pp 701-708.
Livingstone. 16. Runyan CW, Casteel C, Perkis D, et al: Unintentional injuries
8. Granick S, Levere RD: Heme synthesis in erythroid cells. in the home in the United States: Part I. Mortality. Am J Prev
In Moore CV, Brown EB, editors: Progress in hematology, Med 28:7379, 2005.
vol IV, New York, 1964, Grune & Stratton, pp 1-47. 17. Krenzelok EP, Roth R, Full R: Carbon monoxide: the silent killer
9. Dessypris EN, Sawyer ST: Erythropoiesis. In Greer JP, Foerster with an audible solution. Am J Emerg Med 14:484486, 1996.
J, Rodgers GM, et al, editors: Wintrobes clinical hematology, 18. Ryan DH: Examination of the blood. In Lichtman MA, Beutler
ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott, E, Kipps TJ, et al, editors: Williams hematology, ed 7, New York,
Williams & Wilkins, pp 106-125. 2006, McGraw-Hill Professional, pp 11-12.
11 Iron Metabolism
Mary Coleman

OUTLINE OBJECTIVES
Dietary Iron After completion of this chapter, the reader will be able to:
Iron Absorption and
Excretion 1. Discuss the role of iron as an essential nutrient for 6. Describe transferrin, transferrin receptor, hepcidin,
Iron Cycle and Transport human survival. hemosiderin, and ferritin, including function and
Iron Storage 2. List the sites in which iron is distributed in the body regulation.
Laboratory Assessment of and state the approximate amount in each site. 7. Diagram the transport of iron from ingestion to incor-
Iron Metabolism 3. State the minimum daily intake of iron required at poration into heme.
various ages for men, women, and children. 8. Define sideroblast, siderocyte, and siderosome.
4. Describe the mechanism of iron absorption and 9. Discuss regulation, excretion, transport, and storage
distinguish between the absorption of heme and of iron in the body.
nonheme iron. 10. Identify and discuss the laboratory tests currently
5. State the site of iron absorption. used to assess iron status in the body.

CASE STUDY
After studying the material in this chapter, the reader will be able to respond to the following case study:

In 1995, Garry, Koehler, and Simon assessed changes in statistically significant difference in iron stores between the
stored iron in 16 female and 20 male regular blood donors men who took supplements and those who did not, although
aged 64 to 71. They measured Hct, Hb, serum ferritin con a difference was seen for the women. Total iron losses over
centration, and transferrin saturation in samples from the 80 days, the average interval between donations, were calcu
donors, who gave an average of 15 units (approximately lated to be 4.32mg/kg for the women and 3.93mg/kg for
485mL) of blood over 3 1 2 years. The investigators collected the men. As iron stores decreased, the calculated iron absorp
comparable data from nondonors. Of the donors, 10 women tion rose to 3.55mg/day for the women and 4.10mg/day for
and 6 men took a dietary supplement providing approxi the men.
mately 20mg of iron per day. In addition, mean iron dietary 1. Why did the donors iron stores decrease?
intake was 16.4mg/day for the women and 19.9 for the men. 2. What is the average daily iron absorption rate?
Over the period of the study, mean iron stores in women 3. Why did the donors absorption rate rise?
decreased from 12.53 to 1.14mg/kg of body weight. Mean 4. List the laboratory tests performed on serum that are used
iron stores in men declined from 12.45 to 1.92mg/kg. to evaluate iron stores.
Nondonors iron stores remained unchanged. Based on Hb 5. What are the reference intervals for these iron study tests?
and Hct results, no donors became anemic. There was no 6. What does each test result indicate about iron stores?

I
ron in its cationic bivalent ferrous (Fe2+) and trivalent ferric globin chains to produce tetrameric hemoglobin. Ferrous iron
(Fe3+) states is essential for the life of all organisms, plant likewise binds to myoglobin, the O2 transport molecule in
and animal.1 In humans, 70% of total body iron is trans muscles. The primary and secondary structures of myoglobin
ported in blood noncovalently bound in the ferrous state to resemble hemoglobin, however, myoglobin is monomeric,
the heme portion of hemoglobin, where it binds, transports, and and myoglobin oxygen binding is irreversible. Approximately
releases oxygen (O2, Table 11-1).2 In mitochondria, ferrous ion 5% of total human body iron is bound to myoglobin.
is transferred to protoporphyrin IX (see Figure 10-5) to form Ferritin and hemosiderin are the storage forms of iron, con
heme. Four heme molecules become bound, one each, to four taining 25% of human body iron, and are distributed to the

126
CHAPTER 11 Iron Metabolism 127

TABLE 11-1 Iron Compartments in Normal Humans and heme-containing enzymes. Approximately 5% to 35% of
heme iron is absorbed as hemin (iron-containing porphyrin).
Percent of Iron Content
Nonheme iron, found in nonmeat sources such as legumes
Compartment Total Body Iron (mg/kg Body Weight)
and leafy vegetables, accounts for approximately 90% of dietary
Hemoglobin iron 65% 2.6 iron, but only 2% to 20% of it is absorbed, depending on the
Storage iron: 25% 14.2 iron status of the individual and the ratio of dietary enhancers
hemosiderin, ferritin and inhibitors.1 Ascorbate, citrate, and other organic acids and
Myoglobin iron 5% 1.0 amino acids enhance absorption of nonheme iron by the
Transferrin 0.1% 0.003 formation of soluble chelates. Cooking in iron pots increases
Other compartments: 3.6% 0.140 the amount of iron consumed. Substances that interfere with
Peroxidase, catalase, nonheme iron absorption include phytates, polyphenols,
cytochromes, phosphates, oxalates, and calcium.4,5
riboflavin enzymes Dietary iron may be supplemented with tablets or multi
vitamins that supply ferrous sulfate. Since the 1940s, infant
formula and some cereals have been fortified with iron in the
United States. Iron supplementation should be targeted to
liver and bone marrow in hepatocytes and macrophages. populations that are at risk for iron deficiency, because the
Plasma ferritin concentration assays are employed clinically to potential for iron overload exists in individuals with adequate
assess the adequacy of iron stores. Less than 1% of iron is iron status.
transported through plasma in the ferric state bound to trans-
ferrin. Plasma transferrin and transferrin saturation assays may
IRON ABSORPTION AND EXCRETION
be used routinely to diagnose iron deficiency.
Finally, heme-bound iron is essential to the mitochondrial The duodenum and upper jejunum are sites of maximal absorp
cytochrome P-450 (CYP 450) system in all animals. Also known tion of iron. For transport of oxygen in Hb, iron must be in
as cytochrome oxidase, this enzymatic system is bound to the ferrous form (Fe2+). To be absorbed from food, iron must
mitochondrial membranes and supports oxidation-reduction be in the form of heme iron (Fe2+) or converted from ferric
reactions such as the hydroxylation of organic molecules from nonheme iron to the soluble ferrous form by a duodenum-
free oxygen. Cytochrome oxidase iron cycles between the specific cytochrome blike protein, DCYTB.6 Uptake of heme iron
ferrous and ferric state as it facilitates electron transfer. Less occurs on heme carrier protein 1, located on the apical membrane
than 1% of human body iron is located in the CYP 450 system. of the duodenal enterocyte.7 Heme iron binds to the enterocyte
Iron exists only transiently as a free cation; it is normally in the mucosal epithelium and is internalized (Figure 11-1).
bound by or incorporated into various proteins. Because of Here the enzyme heme oxygenase degrades heme to produce
its catalytic properties in single-electron oxidation-reduction ferrous iron, carbon monoxide, and bilirubin-IXa.
reactions, free iron forms oxygen radicals that can damage Ferrous iron is transported across the duodenal epithelium
cellular proteins and lipids. bound to the apical divalent metal transporter 1 (DMT1). The
The concentration of storage and transport iron is con ferrous iron is carried to the basolateral membrane (base
trolled by dietary intake and iron loss through bleeding. Iron and sides of the enterocyte membrane), from which it is
deficiency occurs when there is inadequate intake or excessive exported to the portal circulation, a process mediated by
blood loss, causing anemia. ferroportin, a basolateral transport protein. Ferroportin works
Iron overload results from increased absorption owing to in conjunction with a copper-containing iron oxidase known
genetic predisposition or repeated blood transfusions and pro as hephaestin. Hephaestin may facilitate iron egress by reoxida
duces potentially fatal heart and liver disease. Evolution has tion of ferrous to ferric iron.6 The trivalent (ferric) iron must
given humans a mechanism for absorbing dietary iron effi be bound to transferrin to be transported through the circula
ciently but not for eliminating excess iron effectively. Disorders tion. Some iron remains in the enterocytes as ferritin and
of iron metabolism are discussed in Chapter 19. is released to the circulation over a few hours. Enterocyte-
stored ferritin iron is excreted when the cells are exfoliated
in the stool.
DIETARY IRON
Hepcidin, an antimicrobial peptide produced in the liver,
Although many foods have high iron content, the iron may be seems to act as a negative regulator of intestinal iron absorp
minimally bioavailable. The bioavailability of iron depends on tion. It also suppresses release from macrophages. Hepcidin
its chemical form and the presence of non-iron foods that binds to the ferroportin receptor, causing degradation of fer
promote or inhibit absorption. An average American diet may roportin and trapping iron in the intestinal cells. Hepcidin
provide 10 to 20mg of iron/day, but only 1 to 2mg/day is synthesis rises when transferrin is carrying its maximum capac
absorbed. Iron is absorbed in two forms: heme and nonheme. ity of iron (transferrin saturation of more than 50% in females
Heme-bound iron, mainly from meat, is absorbed more or 60% in males), and diminishes when iron saturation is
efficiently than nonheme, inorganic iron and in a different low.7-9 The role of hepcidin in the anemia of chronic inflamma-
manner.3 Heme iron is present in hemoglobin, myoglobin, tion is discussed in Chapter 19.
128 PART II Hematopoiesis

Body Location Function Type of Iron: Heme or Non-heme

Lumen of duodenum Iron from food Hemoglobin or myoglobin Non-heme iron (Fe3+)
+ chelator (e.g., ascorbate)

Conversion to
absorptive form Hemin DCYTB reductase
Enterocyte Membrane Unknown
mucosal transporter (Fe2+)
membrane Divalent metal transporter
Hemin

Heme oxygenase

Enterocyte Iron stored Fe2+ + bilirubin + CO Fe2+


cytoplasm as ferritin
or
exported
to plasma

Fe2+
Enterocyte Membrane Ferroportin
basolaminal transporter
membrane

Oxidizer Hephaestin

Fe3+
(aPo) Transferrin
Plasma of portal
blood vessels
Carried in Divalent transferrin
plasma to
hepatocytes,
macrophages,
developing
RBCs, etc

Figure 11-1 Simplified version of the mechanism for absorption of iron in the small intestine. aPo, (Apo) transferrin; CO, carbon monoxide;
DCYTB, duodenum-specific cytochrome blike protein; RBC, red blood cell.

Transferrin transports ferric iron (Fe3+) to hematopoietic iron is 1 to 2mg/day. With decreased iron stores, iron absorp
and other tissues, where it is bound by cell membrane transfer- tion may adaptively rise to 3 to 4mg/day. In iron overload,
rin receptors (Figure 11-2). Transferrin receptors are expressed only 0.5mg/day is absorbed.
in larger amounts on normoblasts and rapidly dividing cells, The body conserves iron extremely well, losing only about
whether normal or malignant, but not on highly differentiated one one-thousandth of the total body iron content. This
cells such as reticulocytes.10 Transferrin is taken into the cell by amount is replaced if dietary sources are adequate. Normal
endocytosis. An endosome forms containing the iron-loaded iron losses occur mainly by way of the feces and amount to
transferrin molecule. Iron is released from transferrin by acidi about 1mg/day. Perspiration and exfoliation of the skin and
fication of the endosome to a pH of 5.5. Iron is transported dermal appendages result in minimal losses. Lactation and
across the endosomal membrane by DMT1 and used in the menstruation result in an additional loss of about 1mg/d.11
synthesis of iron-containing proteins. Excess iron is stored as
ferritin or hemosiderin (see section on iron storage).6
IRON CYCLE AND TRANSPORT
Humans have no effective means to excrete iron. Instead we
regulate iron by controlling absorption. The amount of iron Iron is absorbed from the gastrointestinal tract and transported
absorbed is inversely proportional to iron stores and the rate via the circulation to the bone marrow, where it is inserted into
of erythropoiesis. As stated previously, normal absorption of protoporphyrin IX in the mitochondria of the erythroid
CHAPTER 11 Iron Metabolism 129

Location Form of Iron

Plasma Transferrin + Fe3+

Hematopoietic cell Transferrin receptor (TfR) on cell membrane of marrow erythroblasts

Endosome in cell acidified Transferrin + Fe3+ at pH ~ 5.5

Iron released and used to synthesize iron-containing protein such as Hb

Hb production (normoblasts reticulocytes)

Circulation Older reticulocytes and mature RBCs

Macrophages 120-day-old RBCs

Iron + protoporphyrin IX + globin

Plasma Transferrin + Fe3+

Figure 11-2 Cycling of an iron molecule through the body after it has been absorbed in the plasma. Iron is incorporated into marrow red blood cell (RBC)
precursors, released into the circulation for the life of the RBC, and sent back to the plasma, bound to transferrin. Hb, Hemoglobin.

precursors to make heme (refer to Figures 10-2 and 10-5; see The transferrin receptor is a glycoprotein dimer, and it is
also Figure 11-2). Hb synthesis is completed in the reticulocyte located on virtually all cells except mature RBCs. It provides
stage. Iron circulates in red blood cells (RBCs) in the ferrous transferrin-bound iron access into the normoblast; it also
form noncovalently bound to the Hb molecule. Iron from plays a crucial role in the release of iron from transferrin
senescent RBCs is turned over to macrophages and reused.6 within the cell. It is present in large numbers on normoblasts,
Ferrokinetics involve transferrin, transferrin receptor, and in the placenta, and in the liver. The transferrin receptor
ferritin. These are regulated by iron-responsive protein (IRP). can bind two molecules of transferrin. The transferrin recep
The genes for these principal proteins have been located tors affinity for transferrin depends on the iron content of
and sequenced.12 Researchers may someday be able to modify transferrin and the pH. At high levels of transport iron and
these genes to treat hereditary iron disorders. a pH of 7.4, the transferrin receptor selectively binds
Most plasma transferrin is produced by hepatocytes. The diferric transferrin in preference to monoferric transferrin
major function of transferrin is to transport iron from the or apotransferrin. The transferrin receptor gene is located
enterocytes of the duodenum to transferrin receptors on near the transferrin gene at chromosome band 3q26.2-qter
marrow normoblasts. Transferrin has a half-life of 8 days and (see Figure 11-3).12
migrates to the fraction in serum electrophoresis. The amino Control of transferrin receptor biosynthesis is a major
terminus and carboxy terminus each independently bind a mechanism for regulation of iron metabolism. Synthesis is
ferric ion. A bicarbonate ion locks the iron in place within induced by iron deficiency. When transferrin is fully saturated,
transferrin by serving as a bridging ligand between the protein iron is deposited in the liver. When transferrin is congenitally
and iron. The transferrin molecule can exist as a single-chain absent, iron is absorbed by the intestine and accumulates in
glycoprotein with no iron attached, called apotransferrin, or in the liver, pancreas, spleen, and other viscera; only a little makes
a monoferric or diferric form.13 The transferrin gene is located its way to the marrow, and a severe hypochromic microcytic
on the long arm of chromosome 3, at 3q21-qter (Figure 11-3).12 anemia results.12
130 PART II Hematopoiesis

iron (see Chapter 10).6,11 There is evidence that, in addition to


regulation at the mRNA level, transcriptional regulation of iron
metabolism also occurs.11

IRON STORAGE

p Iron is stored in accessible reserve form as ferritin or as the


partially degraded or precipitated form of ferritin called hemo-
siderin. Apoferritin, the protein component of the ferritin
molecule without the iron, is an empty sphere 12nm in dia
meter and 1nm thick and is composed of 24 light (L) and
heavy (H) subunits. Within the apoferritin shell, ferric ions,
hydroxyl ions, and oxygen are distributed in a lattice. The liver
and spleen, which have major iron storage deposits, have a
large amount of L subunits. Tissues such as heart tissue that do
not normally store ferritin have a higher proportion of H sub
units. Genes for the H and L chains belong to multigene fami
lies with members on several chromosomes. The gene for two
types of L chains is on chromosome 19; the gene for heavy
q chains is on chromosome 11.12
Hemosiderin is a degradation product of ferritin produced
by the partial digestion of protein and release of iron micelles,
26.1 Transferrin gene which form insoluble ferritin aggregates. Most stored iron is
3q21-qter soluble ferritin, but as iron stores increase, so does the propor
26.2
Transferrin 26.3 tion of hemosiderin in relation to ferritin.
receptor gene 27 Ferritin and hemosiderin stores are found in the liver, bone
3q26.2-qter 28 marrow, and spleen, with most in the liver. When iron is
29
needed from ferritin iron stores, it is returned to transferrin to
Chromosome 3 be used by the cells that need the iron for metabolism.
Figure 11-3 Location of the transferrin and transferrin receptor genes on
chromosome 3.
LABORATORY ASSESSMENT OF
IRON METABOLISM
Cellular uptake of iron is mediated largely by interaction of Reference intervals for laboratory tests of iron status are pro
the transferrin receptor and the transferrin molecule. The ferric vided but are meant to be a guide only, because values may
irontransferrin receptor complex is endocytosed, iron is differ depending on the assessment method used (Table 11-2).
released into the cell, and the receptor-transferrin complex is A single assay may not provide sufficient iron status informa
returned to the cell surface, where the transferrin is released for tion. An understanding of what the tests measure and what
reuse. Iron enters a chelatable soluble pool in the cell, where variations can occur because of diurnal variation and clinical
it is used for synthesis of essential cellular constituents or for status is helpful in interpreting the significance of the test
deposition as ferritin, a nontoxic storage form of iron. result.
IRPs are messenger ribonucleic acid (mRNA)binding pro Serum iron concentration is a measure of Fe3+ bound to serum
teins that coordinate the intracellular expression of transferrin transferrin and does not include free serum Hb iron. Serum
receptor, ferritin, and other proteins important for iron meta iron concentration is measured using chromogen spectropho
bolism. IRPs bind to iron-responsive elements (IREs), which are tometry. Iron exhibits diurnal variation, being at its highest
stem-loop mRNA structures. IRPs bind IREs when iron supply concentration in the morning and at its lowest in the evening.
is decreased and dissociate from IREs when iron supply is Serum iron concentration is decreased not only in iron
increased. When there is little intracellular iron, IRPs regulate deficiency but also in inflammatory disorders, in acute
the increase of the translation and stability of the mRNA for infection, after certain immunizations, and after myocardial
transferrin receptor and aminolevulinic acid synthase and infarction.
decrease the translation of apoferritin mRNA. This increases Only about one third of transferrin iron-binding sites are
the number of molecules of transferrin receptor on the cell normally occupied by Fe3+; two thirds of the iron-binding sites
membrane, while decreasing the ferritin trapping of iron that do not carry iron, and these sites are referred to collectively as
enters the cell. More Hb can be formed if there is more iron the serum unsaturated iron-binding capacity (UIBC). The total of
uptake by the cell. At the same time, an increase in production available sites is referred to as the total iron-binding capacity
of aminolevulinic acid synthase ensures that enough protopor (TIBC). UIBC is measured using chromogen spectrophotome
phyrin can be made to accommodate the expected increase in try. TIBC can be measured indirectly by chemical means and
CHAPTER 11 Iron Metabolism 131

TABLE 11-2 Assessment of Body Iron Status Serum transferrin receptors (sTfR) can be measured by immu
noassay. The sTfR appears to be a truncated form of the cell
Laboratory Adult Reference
membrane receptor and circulates bound to transferrin. It
Assay (Intervals) Diagnostic Use
seems to mirror the cellular form. The amount of circulating
Serum ferritin level 12-300mcg/L Indicator of iron stores receptor rises when cells lack iron and decreases in chronic
Serum iron level 10-30mol/L Indicator of tissue iron supply diseases. Measurement of sTfR level may be useful in iron
Serum transferrin 47-70mol/L Indicator of tissue iron supply deficiency testing, although increases in levels also have been
level (TIBC) seen with ineffective erythropoiesis.6,11,16 A calculation of the
Transferrin Female: 16-50% Indicator of tissue iron supply ratio of sTfR level to log of serum ferritin concentration (sTfR-F
saturation Male 16-60% index) may be used to differentiate iron deficiency from anemia
Serum transferrin 1.15-2.75mg/L Indicator of functional iron of chronic disease.17
receptor (sTfR) available Erythrocyte protoporphyrin is an intermediate product of
sTfR-F index 0.63-1.8 Indicator of functional iron hemoglobin synthesis that resides in the RBC, and levels may
available be elevated when heme production is incomplete, such as
RBC zinc <80mcg/dL of Indicator of functional iron when iron deficiency is present or when iron use is blocked.
protoporphyrin RBCs available The protoporphyrin assay is termed free erythrocyte protopor
Bone marrow or Normal iron stores Visual qualitative assessment phyrin. Protoporphyrin combines with zinc when iron is
liver biopsy visualized of tissue iron stores unavailable to produce fluorescent zinc protoporphyrin. The
Bone marrow 20-80% Direct assessment of fluorescence of zinc protoporphyrin is measured using a fluo
sideroblast count sideroblasts functional iron available rometer. RBC free protoporphyrin concentration increases in
disorders of heme synthesis, including iron deficiency, lead
RBC, Red blood cell; TIBC, total iron-binding capacity. poisoning, and sideroblastic anemias (see Chapter 19). Proto
porphyrin levels are increased in chronic diseases, regardless of
the iron status.16
Tissue iron is assessed using a biopsy of the bone marrow or
directly by immunoassay.14,15 Direct TIBC measurement is liver. The amount of iron in macrophages, nucleated RBCs, and
really a transferrin assay. reticulocytes is estimated visually using the Prussian blue reac
Measurement of serum iron concentration alone provides tion, which stains iron blue. Iron storage concentration can be
little useful clinical information. To diagnose iron deficiency, estimated by reviewing the amount of iron in macrophages.
serum iron concentration and TIBC are measured and the The Prussian blue stain also can be used on peripheral blood
percent transferrin saturation (the percentage of sites available and bone marrow aspirate smears to visualize iron in nucleated
for carrying iron) is calculated. Percent transferrin saturation is and non-nucleated RBCs. Normally, no iron is detected visu
computed by dividing the serum iron level by the TIBC and ally in mature RBCs or reticulocytes in the peripheral blood.
multiplying the result by 100.6,11,12 In the normal bone marrow smear, one to three blue inclusions/
Ferritin is measured by various immunoassays. The plasma iron granules are found in 20% to 80% of the nucleated RBCs.
ferritin concentration is in equilibrium with ferritin body Nucleated RBCs containing iron cells are called sideroblasts, and
stores. Variations in stored ferritin are reflected in the plasma the granules are called siderosomes. Reticulocytes in the bone
ferritin concentration. The plasma ferritin concentration marrow that contain iron are termed siderocytes. The presence
declines early in the development of iron deficiency. As an of siderocytes is an abnormal finding, because the iron
acute phase reactant, however, it is increased in inflammation, granules in reticulocytes should not be detectable at the light
regardless of the quantity of iron in storage. microscope level.

SUMMARY
Iron is an essential nutrient for human survival. It plays a role in process by reoxidation of ferrous iron to ferric iron. Hepcidin, an
oxygen transport and basic metabolic oxidation-reduction reac- antimicrobial peptide, regulates the basolateral iron export.
tions. Most of the iron in the body is in the form of hemoglobin. Iron is transported in plasma bound to a carrier protein, transferrin.
Iron is obtained through the diet via heme (Fe2+) and nonheme Transferrin receptors, located on all cells in the body except mature
(inorganic Fe3+) iron. Heme is more readily absorbed than RBCs, aid in providing transferrin-bound iron access into cells and
nonheme iron. play a crucial role in the release of iron from transferrin within the
Iron is absorbed by duodenum and jejunum enterocytes via the cell. The IRP regulates aminolevulinic acid synthetase, transferrin
DMT1 transporter. Iron is carried to the basolateral membrane, receptors, and ferritin in the cell by binding to the mRNA IREs.
where it passes to the plasma via a channel transporter, ferropor- Ferritin and hemosiderin are storage forms of iron. Most stored
tin. Hephaestin, a copper oxidase protein, aids in the transport iron is in the form of ferritin, which is a water-soluble complex.
132 PART II Hematopoiesis

Hemosiderin, a water-insoluble complex, is composed of ferritin protoporphyrin test. Other tests are available but are performed
aggregates and is found in macrophages in the bone marrow and less frequently.
liver.
Laboratory tests that can assist in assessing the iron status in Now that you have completed this chapter, go back and
the body include serum ferritin level, serum iron concentration, read again the case study at the beginning and respond
TIBC, and percent transferrin saturation. Other tests include to the questions presented.
bone marrow or liver biopsy, sTfR analysis, and the erythrocyte

R E V I E W Q UESTIONS
1. Iron is transported in plasma via: 7. Below are several of the many steps in the process from
a. Hemosiderin absorption and transport of iron to incorporation into
b. Ferritin heme. Place them in proper order.
c. Transferrin i. Transferrin picks up ferric iron.
d. Hemoglobin ii. Iron is transferred to the mitochondria.
iii. Cytochrome blike protein, DCYTB, converts ferric
2. What is the major metabolically available storage form of to ferrous iron.
iron in the body? iv. Ferroportin transports iron across the cell
a. Hemosiderin membrane.
b. Ferritin v. The transferrin receptor transports iron into
c. Transferrin the cell.
d. Hemoglobin a. v, iv, i, ii, iii
b. iii, ii, iv, i, v
3. Approximately 70% of body iron is found in the form of: c. ii, i, v, iii, iv
a. Hemosiderin d. iii, iv, i, v, ii
b. Ferritin
c. Transferrin 8. In the iron cycle, the transferrin receptor does which of the
d. Hemoglobin following?
a. Carries iron into duodenal cells from the intestinal
4. Iron is incorporated into the heme molecule in which of lumen
the following forms? b. Carries iron out of duodenal cells into the plasma
a. Ferro c. Carries transferrin-bound iron in the plasma
b. Ferrous d. Carries transferrin-bound iron into erythroid
c. Ferric precursors
d. Apoferric
9. What is the fate of the transferrin receptor when it has
5. What protein associated with duodenal cells transports completed its role in the delivery of iron into a developing
iron from the intestinal lumen into the intestinal cell? RBC?
a. Transferrin a. It is recycled to the plasma membrane and released
b. Ferroportin into the plasma.
c. DMT1 b. It is recycled to the plasma membrane, where it can
d. Ferrochelatase bind its ligand again.
c. It is catabolized and the amino acids are returned to
6. Iron is transported out of cells such as macrophages and the metabolic pool.
duodenal cells by what membrane protein?
a. Transferrin 10. The transfer of iron from the duodenal cell into the plasma
b. Ferroportin is regulated by:
c. DMT1 a. Transferrin
d. Ferrochelatase b. Transferrin receptor
c. Hephaestin
d. Hepcidin
CHAPTER 11 Iron Metabolism 133

REFERENCES
1. Beard JL, Dawson H, Pinero DJ: Iron metabolism: a compre 10. Ponka P, Lok CN: The transferrin receptor: role in health and
hensive review. Nutr Rev 54:295-317, 1996. disease. Int J Biochem Cell Biol 31:1111-1137, 1999.
2. Sharma N, Butterworth J, Cooper BT, et al: The emerging role 11. Beutler E: Disorders of iron metabolism. In Lichtman
of the liver in iron metabolism. Am J Gastroenterol 100:201- MA, Beutler E, Kipps, et al, editors: Williams hematology,
206, 2005. ed 7, New York, 2006, McGraw-Hill Professional, pp 511-
3. Uzel C, Conrad ME: Absorption of heme iron. Semin Hematol 553.
5:27-34, 1998. 12. Rouault TA, Klausner RD: Molecular basis of iron metabo
4. Conrad ME: Introduction: iron overloading and iron regula lism. In Stamatoyannopoulos G, Majerus PW, Perlmutter RM,
tion. Semin Hematol 35:1-4, 1998. et al, editors: Molecular basis of blood diseases, ed 3,
5. Andrews NC: Iron deficiency and related disorders. In Philadelphia, 2001, Saunders, pp 363-387.
Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes 13. Woods S, DeMarco T, Friedland M: Iron metabolism. Am J
clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer Gastroenterol 85:1-8, 1990.
Health/Lippincott Williams & Wilkins, pp 810-830. 14. Worwood M: The laboratory assessment of iron statusan
6. Andrews NC: Pathology of iron metabolism. In Hoffman R, update. Clin Chim Acta 259:3-23, 1997.
Benz EJ, Shattil SJ, et al, editors: Hematology, basic principles 15. Higgins T, Beutler E, Doumas BT: Hemoglobin, iron and
and practice, ed 5, New York, 2009, Elsevier. Available at: bilirubin. In Burtis CA, Ashwood ER, Bruns DE, editors:
http://www.mdconsult.com/book/player/book.do?method= Tietz textbook of clinical chemistry and molecular diagnostics,
display&type=aboutPage&decorator=header&eid=4-u1.0-B978- ed 4, Philadelphia, 2006, Saunders, pp 1165-1204.
0-443-06715-0..X5001-8--TOP&isbn=978-0-443-06715-0& 16. Gardner LB, Benz EJ: Anemia of chronic disease. In Hoffman
uniq=175816539#lpState=open&lpTab=contentsTab& R, Benz EJ, Shattil SJ, et al, editors: Hematology, basic principles
content=4-u1.0-B978-0-443-06715-0..50037-6%3Bfrom% and practice, ed 5, New York, 2009, Elsevier. October 2, 2009.
3Dtoc%3Btype%3DbookPage%3Bisbn%3D978-0-443 Available at: http://www.mdconsult.com/book/player/book.
-06715-0. Access date October 2, 2009. do?method=display&type=aboutPage&decorator=header&
7. Bleackley MR, Wong AYK, Hudson DM, et al: Blood iron eid=4-u1.0-B978-0-443-06715-0..X5001-8--TOP&isbn=978-0-
homeostasis: newly discovered proteins and iron balance. 443-06715-0&uniq=175816539#lpState=open&lpTab=
Transfusion Med Rev 23:103-123, 2009. contentsTab&content=4-u1.0-B978-0-443-06715-0..50039-
8. Andrews NC: Forging a field: the golden age of iron biology. X%3Bfrom%3Dtoc%3Btype%3DbookPage%3Bisbn%
Blood 112:219-230, 2008. 3D978-0-443-06715-0. Access date October 2, 2009.
9. Anderson GJ, Frazer DM, McLaren GD: Iron absorption and 17. Muoz M, Villar I, Garca-Erce JA: An update on iron physio
metabolism. Curr Opin Gastroenterol 25:129-135, 2009. logy. World J Gastroenterol 15:4617-4626, 2009.
12 LeukocyteandDevelopment,
Functions
Kinetics,

Anne Stiene-Martin

OUTLINE OBJECTIVES
Granulocytes After completion of this chapter, the reader will be able to:
Neutrophils
Eosinophils 1. Describe the Wright stain morphology of neutrophils 6. Compare the kinetics of neutrophils and monocytes.
Basophils at the five stages of maturation and name each stage. 7. Discuss in general terms how the various types of
Mast Cells 2. Describe the Wright stain morphology of immature lymphocytes are produced and describe the Wright
Mononuclear Cells and mature eosinophils and basophils. stain morphology of resting lymphocytes, immature B
Monocytes 3. Discuss the various types of granules found in neutro- cells, effector B cells, and cytotoxic T cells or natural
Lymphocytes phils, eosinophils, and basophils and list at least three killer cells.
compounds found in the granules of each cell line. 8. Define what is meant by the term blastogenesis as
4. Describe in general terms the activity known as related to lymphocyte activation.
phagocytosis. 9. Describe at least three functions of each type of
5. Describe the Wright stain morphology of promono- leukocyte covered in this chapter.
cytes, monocytes, and macrophages.

L
eukocytes (also known as white blood cells or WBC) are whose nuclei are not segmented but usually round, oval,
so named because they are relatively colorless compared indented, or folded. These two types of leukocytes are called
with red blood cells. The number of different types of mononuclear cells. Mononuclear cells can be subdivided into
leukocytes varies depending on whether they are being viewed monocytes and lymphocytes. The manner in which the hetero-
with a light microscope after staining with a Romanowsky stain chromatin (see Chapter 6) is clumped is referred to as the
(five or six types) or are identified according to their surface chromatin pattern and is relatively unique for some of the leu-
antigens using flow cytometry (at least ten different types). kocytes. This fact aids in their identification, and this is espe-
For the purposes of this chapter, the classic, light microscope cially true when lymphocytes and monocytes are compared.
classification of leukocytes will be used. Leukocytes develop from primitive stem cells in the bone
The names given to the individual types of leukocytes arise marrow, where most undergo differentiation and maturation
from a combination of their acid-base staining characteristics (Figure 12-1), and then are released into the circulation. The
and their nuclear shape and character. Thus there are three number of circulating leukocytes varies with sex, age, activity,
different types of leukocytes whose cytoplasm is filled with time of day, and ethnicity; it also differs according to whether
granules with differing staining characteristics and whose or not the leukocytes are reacting to stress, being consumed, or
nuclei are segmented or lobulated. As a group, they are referred being destroyed, and whether or not they are being produced
to as granulocytes. Individually, they are known as eosinophils, by the bone marrow in sufficient numbers.1 Reference ranges
with granules containing basic proteins that stain with acid for total leukocyte counts vary among laboratories depending
stains such as eosin; basophils, with granules that are acidic and on the patient population (e.g., a pediatric hospital compared
stain with basic stains such as methylene blue; and neutrophils, to a geriatric clinic) and the type of instrumentation being used.
with granules that react with both acid and basic stains, which Generally speaking, the range can be as low as 3.8 109/L and
gives them a pink to lavender color. Because nuclear segmenta- as high as 25 to 30 109/L for infants or 11.5 109/L for adults.
tion is quite prominent in neutrophils, they have also been The overall function of leukocytes is protection from foreign
called polymorphonuclear cells or PMNs. organisms, cells, or material. This is referred to as immunity and
In addition to the three granulocytes, there are two other may be nonspecific (innate), as in phagocytosis by neutrophils
cell types whose cytoplasm may or may not contain visible or monocytes, or specific (adaptive), as in the production of
granules that stain with azures (azurophilic granules) and specific antibodies by lymphocytes. The term kinetics refers to

134
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 135

Long-term self- Short-term self-


renewing stem cell renewing stem cell

Multipotent progenitor
hematopoietic stem cell

Common Common
myeloid progenitor lymphoid progenitor

Granulocyte-monocyte Eosinophil-basophil Megakaryocyte-erythrocyte Dendritic Pre-B Pre-T Natural


progenitor progenitor progenitor cell killer cell

Myeloblast Monoblast Myeloblast Myeloblast Pronormoblast Megakaryoblast B lymphoblast T lymphoblast

Neutrophil Monocyte Eosinophil Basophil Erythrocyte Megakaryocyte B cell T cell

Macrophage Mast cell Platelets Plasma cell


Figure 12-1 Hematopoietic chart representing the derivation pathways of each type of blood cell.

the movement of cells through developmental stages, into the in relative numbers and 2.3 to 8.7 109/L in absolute terms).
circulation, and from the circulation to the tissues and includes As can be seen from the table on the inside front cover, pedi-
the time spent in each phase of the cells life. As each cell type atric values are quite different; relative percentages can be as
is discussed in this chapter, developmental stages, kinetics, and low as 18% to 20% of leukocytes in the first few months of life
specific functions will be addressed. and do not begin to climb to adult values until after 4 to 7
years of age.

GRANULOCYTES
Neutrophil Development
Neutrophils Neutrophil development occurs in the bone marrow under
Neutrophils are present in the peripheral blood in two forms normal circumstances. Neutrophils share a common progeni-
according to whether the nucleus is segmented (polymorpho- tor with monocytes known as the granulocyte-monocyte progeni-
nuclear or PMN) or still in a band shape. PMNs make up the tor (GMP). The major cytokine responsible for the stimulation
vast majority of circulating neutrophils, with band forms of neutrophil production is granulocyte colony-stimulating
reaching only 3% to 15% of leukocytes depending on identi- factor, or G-CSF.2 Neutrophil development is also under the
fication criteria. Neutrophils are present in the highest numbers control of specific transcription factors and apoptotic regula-
in the peripheral blood of adults (50% to 75% of leukocytes tors that are beyond the scope of this chapter.
136 PART II Hematopoiesis

Figure 12-2 Diagrammatic representation of neutrophil maturation showing stimulating cytokines and the two marrow pools.

There are two pools of developing neutrophils in the


bone marrow (Figure 12-2). The mitotic (or proliferating) pool BOX 12-1 Neutrophil Granules
consists of cells that are dividing by mitosis and includes
hematopoietic stem cells (HSCs)3; common myeloid progeni- Primary (Azurophilic) Granules
tors (CMPs), also known as colony-forming unitsgranulocyte, Formed during the promyelocyte stage
erythrocyte, monocyte, and megakaryocyte (CFU-GEMMs); Last to be released (exocytosis)
GMP4; myeloblasts; promyelocytes; and myelocytes. The Contain:
second marrow pool of neutrophils is the storage (or matura- Myeloperoxidase
tion) pool consisting of cells that are no longer dividing but Acid -glycerophosphatase
are undergoing nuclear maturation that is visible as nuclear Cathepsins
shape changes from round to indented to banded to segmented Defensins
as well as increased clumping of the nuclear chromatin. The Elastase
storage pool consists of metamyelocytes, band form neutro- Proteinase-3
phils, and segmented neutrophils. Others
HSCs, CMPs, and GMPs are not distinguishable with the
light microscope and Romanowsky staining and may resemble Secondary (Specific) Granules
early type I myeloblasts or lymphoid cells. They can, however, Formed during myelocyte and metamyelocyte stages
be identified through surface antigens and flow cytometry Third to be released
(see Chapter 33). The HSC is CD34+CD38-CD133+Thy-1+; Contain:
the CMP is CD34+CD38lowIL3RlowCD45RA;4 and the GMP is 2-Microglobulin
CD34+CD38+CD45RA+.5,6 Collagenase
Myeloblasts make up 1% to 2% of the nucleated cells in the Gelatinase
bone marrow. They are frequently subdivided into types I and Lactoferrin
II. Myeloblasts vary in size depending on the stage of the Neutrophil gelatinase-associated lipocalin
mitotic cycle (14 to 20m). The nucleus is round to oval and Others
usually centrally located. It is homogeneous with delicate
euchromatin and two to four visible nucleoli. The cytoplasm
Tertiary Granules
Formed during metamyelocyte and band stages
is slightly basophilic and the nucleus-to-cytoplasm (N:C) ratio
Second to be released
is 1:1 to 1:0.5 depending on the stage of the mitotic cycle.
Contain:
Type I myeloblasts have no visible granules when stained with
Gelatinase
Romanowsky stains and viewed with the light microscope.
Collagenase
When most of these cells are viewed with the electron micro-
Lysozyme
scope or stained for peroxidase enzyme, granules are apparent.
Acetyltransferase
Those that do not have detectable granules may represent
2-Microglobulin
earlier stem cells such as the CMP. Type II myeloblasts contain
up to 20 visible azurophilic granules starting at the Golgi appa-
Secretory Granules (Secretory Vesicles)
ratus and spreading throughout the cell (Figure 12-3). These
Formed during band and segmented stages
azure granules are referred to variously as primary granules or
First to be released (fuse to plasma membrane)
nonspecific granules because they are the first to appear and they
Contain (attached to membrane):
are not unique to any one cell type.
CD11b/CD18
Promyelocytes make up 1% to 6% of the nucleated cells in
Alkaline phosphatase
the bone marrow. They vary in size depending on the stage of
Vesicle-associated membrane-2
the mitotic cycle (16 to 25m). The nucleus is round to oval
CD10, CD13, CD14, CD16
and is often eccentric. The cytoplasm is evenly basophilic and
Cytochrome b558
full of primary azurophilic granules. These granules are the first
Complement 1q receptor
in a series of granules to be produced during neutrophil matu-
Complement receptor-1
ration (Box 12-1).7 The nucleus is similar to that described
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 137

Rights were not granted to include this figure


in electronic media.
Please refer to the printed publication.

B
Figure 12-4 A, Progranulocyte or promyelocyte. Note the large number
of azure granules and the presence of nucleoli. B, Electron micrograph of a
promyelocyte. (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
Philadelphia, 2009, Saunders.)

division (mitosis) occurs. During this stage, the production of


primary granules ceases, and the cell begins to manufacture sec-
ondary or specific neutrophil granules. This stage of neutrophil
development is sometimes divided into early and late myelo-
cytes. Early myelocytes may look very similar to the promyelo-
cytes described earlier in size and nuclear characteristics except
that patches of grainy pale pink cytoplasm representing second-
ary granules begin to be evident in the area of the Golgi apparatus.
This has been referred to as the dawn of neutrophilia. Secondary
C
neutrophilic granules slowly spread through the cell until its
Figure 12-3 A, Type I myeloblast (arrow). Note that no granules are cytoplasm is more lavender-pink than blue. As the cell divides,
visible in the cytoplasm. B, Type II myeloblast (arrow) with a few azure the number of primary granules per cell is decreased, and their
granules in the cytoplasm. C, Electron micrograph of a myeloblast. (B cour- membrane chemistry changes so that they are much less visible.
tesy Linda M. Marler and Jean A. Sider, Indiana Pathology Image Atlas Series, Late myelocytes are somewhat smaller than promyelocytes (15
Hematology Image Atlas; C from Carr JH, Rodak BF: Clinical hematology
to18m), and the nucleus has considerably more heterochroma-
atlas, ed 3, Philadelphia, 2009, Saunders.)
tin. Nucleoli are difficult to see by light microscopy (Figure 12-5).
Neutrophil metamyelocytes constitute 20% to 30% of nucle-
earlier for myeloblasts except that chromatin clumping (het- ated marrow cells. From this stage forward the cells are no
erochromatin) may be visible, especially around the edges of longer capable of division, and the major morphologic change
the nucleus. Nucleoli are easily seen (Figure 12-4). is in the shape of the nucleus. Synthesis of tertiary granules
Neutrophil myelocytes make up 6% to 17% of the nucleated (also known as gelatinase granules) may begin during this stage.
cells in the bone marrow and are the final stage in which cell The size of the metamyelocyte is a little smaller than that of
138 PART II Hematopoiesis

A
A

B
B
Figure 12-6 A, Two neutrophil metamyelocytes (arrows). Note that there
is no remaining basophilia in the cytoplasm, and the nucleus is indented.
B, Electron micrograph of a neutrophil metamyelocyte. (B from Carr JH,
Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)

during this stage. Secretory granules (also known as secretory


vesicles) may begin to be formed during this stage. The nucleus
is highly clumped, and the nuclear indentation that began in
the metamyelocyte stage now exceeds one half the width of the
nucleus, but actual segmentation has not yet occurred. Over
C the past 70 years there has been considerable controversy over
Figure 12-5 A, Two early neutrophil myelocytes. Note that they are very the definition of a band form and the differentiation between
similar to the progranulocyte except for several light areas in their cytoplasm band forms and segmented forms. There have been up to three
where specific granules are beginning to appear. B, Arrows are pointing to schools of thought from the most conservativeholding that
three late myelocytes in the field. Their cytoplasm has few if any primary a band must have an equal diameter throughout (reference
granules, and the pink secondary granules are easily seen. C, Electron range, 0% to 3% in the peripheral blood)to the most
micrograph of a late neutrophil myelocyte. (C from Carr JH, Rodak BF: Clinical liberalrequiring that a filament between segments be visible
hematology atlas, ed 3, Philadelphia, 2009, Saunders.) before a band becomes a segmented neutrophil (reference
range, 8% to 18% in the peripheral blood). The middle ground
the myelocyte (14 to 16m). The cytoplasm contains very states that when doubt exists, the cell should be called a seg-
little residual ribonucleic acid (RNA) and therefore little or no mented neutrophil (reference range, 5% to 10% in the periph-
basophilia. The nucleus is indented (kidney bean shaped or eral blood) (Figure 12-7). In the past decade, an increasing
peanut shaped), and the chromatin is increasingly clumped. number of laboratories are no longer distinguishing between
Nucleoli are absent (Figure 12-6). band forms and segmented forms, because the clinical utility
Neutrophil band forms make up 9% to 32% of nucleated of this differentiation has been called into question.8
marrow cells. All evidence of RNA (cytoplasmic basophilia) PMN neutrophils (segmented neutrophils) make up 7% to 30%
should be absent, and tertiary granules continue to be formed of nucleated cells in the bone marrow. Secretory granules
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 139

A
A

B
B Figure 12-8 A, Segmented neutrophil (also known as a polymorphonu-
clear cell or PMN). B, Electron micrograph of a segmented neutrophil.
Figure 12-7 A, Band form neutrophil with a nucleus whose indentation (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3, Philadelphia,
is more than 50% of the width of the nucleus. B, Electron micrograph of a 2009, Saunders.)
band neutrophil. (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
Philadelphia, 2009, Saunders.)
50:50 overall10; however, marginated neutrophils in the capil-
continue to be formed during this stage. The only morphologic laries of the lungs make up a considerably larger portion of
difference between this cell and the band form is the presence peripheral neutrophils.11 There do not appear to be any func-
of between three and four nuclear segments connected by tional differences between neutrophils of either the CNP or the
threadlike filaments (Figure 12-8). MNP, and cells move freely between the two peripheral pools.12
The half-life of neutrophils in the blood is relatively short at 6
Neutrophil Kinetics to 8 hours.8 Integrins and selectins are of significant impor-
Neutrophil kinetics involves the movement of neutrophils tance in allowing neutrophils to marginate as well as exit the
between five areas known as pools: the mitotic pool in the bone blood and enter the tissues by a process known as diapedesis.13
marrow, the storage pool in the bone marrow, the circulating Those neutrophils that do not migrate into the tissues eventu-
pool in the peripheral blood, the marginated pool in the ally undergo programmed cell death or apoptosis and are
peripheral blood, and the tissue pool. Neutrophil production removed by macrophages in the spleen.14
has been calculated to be on the order of between 0.9 and Once neutrophils are in the tissues, their life span is variable
1.0 109 cells/kg per day.9 depending on whether or not they are responding to infectious
The mitotic pool contains approximately 2.11 109 cells/ or inflammatory agents. In the absence of infectious or inflam-
kg, whereas the storage pool contains roughly 5.6 109 cells/ matory agents, the neutrophils life span is measured in hours.
kg or a 5-day supply. The transit time from the HSC to the Some products of inflammation and infection tend to prolong
myeloblast has not been measured. The transit time from the neutrophils life span through antiapoptotic signals,
myeloblast through myelocyte has been estimated to be whereas others such as MAC-1 trigger the death and phagocy-
roughly 6 days, and the transit time through the storage pool tosis of neutrophils that have reached the end of their useful
is roughly 6 to 7 days.9 Granulocyte release from the bone lifespan.13
marrow is stimulated by G-CSF.2
Once in the peripheral blood, neutrophils are divided ran- Neutrophil Functions
domly into a circulating neutrophil pool (CNP) and a marginal Neutrophils are part of the innate immune system. Character-
neutrophil pool (MNP). The ratio of these two pools is roughly istics of innate immunity include the following: (1) the
140 PART II Hematopoiesis

destruction of foreign organisms is not antigen specific; (2) it


provides no protection against reexposure to the same patho- BOX 12-2 Phagocytosis
gen; (3) it relies on the barriers provided by skin and mucous
membranes as well as phagocytes such as neutrophils and Recognition and Attachment
monocytes; and (4) it includes a humoral component known Phagocyte receptors recognize and bind to certain foreign
as the complement system. molecular patterns and opsonins such as antibodies and comple-
The major function of neutrophils is phagocytosis and ment components.
destruction of foreign material and microorganisms. The
process involves seeking (chemotaxis, motility, and diapede- Ingestion
sis) and destruction (phagocytosis and digestion). Pseudopodia are extended around the foreign particle and enclose
Neutrophil recruitment to an inflammatory site begins it within a phagosome (engulfment).
when chemotactic agents bind to neutrophil receptors. Che- The phagosome is pulled toward the center of the cell by polym-
motactic agents may be produced by microorganisms, by erization of actin and myosin and by microtubules.
damaged cells, or by other leukocytes such as lymphocytes
or other phagocytes. The first neutrophil response is to roll Killing and Digestion
along endothelial cells using stronger adhesive molecules than Oxygen Dependent
those used by nonstimulated marginated neutrophils. Rolling Respiratory burst through the activation of NADPH oxidase. H2O2 and
consists of transient adhesive contacts between neutrophil hypochlorite are produced.
selectins and adhesive molecules on the surface of endothelial
cells. At the same time, secretory granules containing addi- Oxygen Independent
tional adhesive molecules are fused to the neutrophils plasma The pH within the phagosome becomes alkaline and then neutral, the
membrane. 2 integrins such as CD11b/CD18 from secretory pH at which digestive enzymes work.
granules contribute to tight stationary binding between neu- Primary and secondary lysosomes (granules) fuse to the phagosome and
trophils and endothelial cells. This is followed by diapedesis empty hydrolytic enzymes and other bacteriocidal molecules into the
or transmigration of neutrophils either between or through phagosome.
endothelial cellsa process that is also mediated by integrins
and integrin-associated proteins. Tertiary granules containing Formation of Neutrophil Extracellular Traps
gelatinase and collagenase are released by transmigrating Nuclear and organelle membranes dissolve, and activated cytoplas-
neutrophils. Gelatinase degrades denatured collagen as well as mic enzymes attach to DNA.
types IV and V collagen and activates chemokines such as The cytoplasmic membrane ruptures and DNA with attached
interleukin-8 (IL-8).15 Neutrophils then migrate in a direc- enzymes is expelled, so that the bacteria are digested in the external
tional manner toward the area of greatest concentration of environment.
chemotactic agents. NADPH, Nicotinamide adenine dinucleotide phosphate (reduced form).
Once at the site of infection or inflammation, neutrophils
begin the process of phagocytosis (Box 12-2). They utilize their
enormous inventory of surface receptors either to directly rec-
ognize the pathogen, apoptotic cell, or particle or to recognize for macrophages to phagocytize dead neutrophils as well as
opsonic molecules attached to the foreign particle such as anti- inflammatory agents that may cause tissue damage.
bodies or complement components. With recognition comes A second function of neutrophils is the generation of neu-
attachment and engulfment, in which cytoplasmic pseudopo- trophil extracellular traps or NETs.18 NETs are extracellular
dia surround the particle, forming a phagosome within the threadlike structures believed to represent chains of nucleo-
neutrophil cytoplasm.16 Formation of the phagosome allows somes from unfolded nuclear chromatin material (DNA).
the reduced nicotinamide adenine dinucleotide (NADH) These structures have enzymes from neutrophil granules
oxidase complex within the phagosome membrane to assem- attached to them and have been shown to be able to trap
ble; this leads to the generation of reactive oxygen species such and kill gram-positive and gram-negative bacteria as well as
as hydrogen peroxide, which is converted to hypochlorite by fungi. NETs are generated at the time that neutrophils die as a
myeloperoxidase. Likewise, a series of metabolic changes cul- result of antibacterial activity.
minate in the fusion of primary and/or secondary granules to A third and final function of neutrophils is their secretory
the phagosome and the release of numerous bacteriocidal function. Neutrophils are a source of transcobalamin I or R
molecules into the phagosome.17 This combination of reactive binder protein, which is necessary for the proper absorption of
oxygen species and nonoxygen-dependent mechanisms is vitamin B12. In addition, they are a source of a variety of
generally able to destroy most pathogens. cytokines.
In addition to emptying their contents into phagosomes,
secondary and primary granules may fuse to the plasma mem- Eosinophils
brane, which results in release of their contents into the extra- Eosinophils make up between 1% and 5% of peripheral blood
cellular matrix. These molecules can then act as chemotactic leukocytes, with an absolute number of between 40 and 550
agents for additional neutrophils and as stimulating agents per microliter of blood.
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 141
Eosinophil Development
Eosinophil development is similar to that described earlier for
neutrophils; however, a question remains regarding the imme-
diate progenitors. There is some agreement that a common
precursor exists for eosinophils and basophils, but whether this
common precursor arises from the GMP or from the common
myeloid progenitor (CMP) is still unresolved. The evidence for
a common precursor includes the following: (1) the granules
of both eosinophils and basophils contain major basic protein
and lysophospholipase; (2) both have receptors for IL-3, IL-5
and granulocyte-macrophage colony-stimulating factor (GM-
CSF); (3) individuals with increased basophils often have
A
increased eosinophils; (4) there is a rare syndrome character-
ized solely by the absence of both cell types; (5) cells with
both eosinophil and basophil granules are sometimes seen
in patients with myeloproliferative syndromes; and (6) hybrid
eosinophil-basophil cells have been grown in culture.19
Eosinophil lineage is established through the interaction
between the cytokines IL-3, IL-5, and GM-CSF and three tran-
scription factors (GATA-1, PU.1 and c/EBP). Whether or not
there exist myeloblasts that are committed to the eosinophil
line has not been established. Eosinophilic promyelocytes can
be identified cytochemically due to the presence of Charcot-
Leyden crystal protein in their primary granules. The first B
maturation phase that can be identified as eosinophilic Figure 12-9 A, Eosinophil myelocyte. Note the rounded nucleus and the
using light microscopy and Romanowsky staining is the blue background cytoplasm in which there are numerous large, pale eosino-
early myelocyte. phil granules. B, Electron micrograph of eosinophil granules showing the
Eosinophil myelocytes are characterized by the presence of central crystalline core in some of the granules. (B from Carr JH, Rodak BF:
large (resolvable at the light microscope level), pale, reddish Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
orange secondary granules along with azure granules in blue
cytoplasm. The nucleus is similar to that described for neutro-
phil myelocytes. Transmission electron micrographs of eosino-
phils reveal the fact that many secondary eosinophil granules
contain an electron-dense crystalline core (Figure 12-9).20
Eosinophil metamyelocytes and band forms resemble their
neutrophil counterparts with respect to their nuclear shape.
Secondary granules increase in number, and a third type of
granule is generated called the secretory granule or secretory
vesicle. The secondary granules become more distinct and
refractory. Electron microscopy indicates the presence of two
other organelles: lipid bodies and small granules (Box 12-3).21
Mature eosinophils differ from segmented neutrophils in that
their nuclei generally have only two segments. The cytoplasm
is full of the characteristic eosinophilic secondary granules
(Figure 12-10). Electron microscopy of mature eosinophils Figure 12-10 Mature eosinophil. Note that the nucleus has only two
reveals secretory vesicles to be extensive, and their number segments, which is usual for these cells. The background cytoplasm is color-
increases considerably when the eosinophil is stimulated or less and filled with eosinophil secondary granules. (From Carr JH, Rodak BF:
activated.20 Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)

Eosinophil Kinetics
Approximately 3% of nucleated bone marrow cells are eosino- of eosinophils in the marrow consisting of between 9 and
phils. Of these, slightly more than a third are mature, a quarter 14 108 cells/kg.21
are metamyelocytes, and the remainder are promyelocytes or Once in the circulation, eosinophils have a circulating
myelocytes. The time from the last myelocyte mitotic division half-life of roughly 18 hours22; however, the half-life of eosino-
to the emergence of mature eosinophils from the marrow is phils is prolonged when eosinophilia occurs. The tissue desti-
about 3.5 days. The mean turnover of eosinophils is approxi- nations of eosinophils under normal circumstances appear to
mately 2.2 108 cells/kg per day. There is a large storage pool be underlying columnar epithelial surfaces in the respiratory,
142 PART II Hematopoiesis

tents into the extracellular space. Compound exocytosis


BOX 12-3 Eosinophil Granules is a second mechanism in which granules fuse together
within the eosinophil prior to fusing with the plasma mem-
Primary Granules brane. A third method is known as piecemeal degranulation,
Formed during promyelocyte stage in which secretory vesicles remove specific proteins from the
Contain: secondary granules. These vesicles then migrate to the plasma
Charcot-Leyden crystal protein membrane and fuse to empty the specific proteins into the
extracellular space.24
Secondary Granules Eosinophils play important roles in immune regulation.
Formed throughout remaining maturation They transmigrate into the thymus of the newborn and are
Contain: believed to be involved in the deletion of double-positive
Major basic protein (core) thymocytes.25 Eosinophils are capable of acting as antigen-
Eosinophil cationic protein (matrix) presenting cells and promoting the proliferation of effector T
Eosinophil-derived neurotoxin (matrix) cells.26 They are also implicated in the initiation of either type
Eosinophil peroxidase (matrix) 1 or type 2 immune responses due to their ability to rapidly
Lysozyme (matrix) secrete preformed cytokines in a stimulus-specific manner.27
Catalase (core and matrix) Eosinophils regulate mast cell function through the release of
-Glucuronidase (core and matrix) major basic protein that causes mast cell degranulation as well
Cathepsin D (core and matrix) as cytokine production, and they also produce nerve growth
Interleukins 2, 4, and 5 (core) factor that promotes mast cell survival and activation.
Interleukin-6 (matrix) Eosinophil production is increased in infection by parasitic
Granulocyte-macrophage colony-stimulating factor (core) helminths, and in vitro studies have shown that the eosinophil
Others is capable of destroying tissue-invading helminths through
the secretion of major basic protein and eosinophil cationic
Small Lysosomal Granules protein as well as the production of reactive oxygen species.26
Acid phosphatase There is also a suggestion that they play a role in preventing
Arylsulfatase B reinfection.28
Catalase Finally, eosinophilia is a hallmark of allergic disorders, of
Cytochrome b558 which asthma has been the best studied. The number of eosin-
Elastase ophils in the blood as well as in sputum correlates with disease
Eosinophil cationic protein severity. This has led to the suggestion that the eosinophil is
one of the causes of airway inflammation and mucosal cell
Lipid Bodies damage through secretion or production of a combination of
Cyclooxygenase basic proteins, lipid mediators, and reactive oxygen species.26
5-Lipoxygenase Eosinophils have also been implicated in airway remodeling
15-Lipoxygenase through eosinophil-derived fibrogenic growth factors.29 Eosin-
Leukotriene C4 synthase ophil accumulation in the gastrointestinal tract occurs in
Eosinophil peroxidase allergic disorders such as food allergy, allergic colitis, and
Esterase inflammatory bowel disease.30

Storage Vesicles Basophils


Carry proteins from secondary granules to be released into the extracel- Basophils and mast cells are two cells with morphologic and
lular medium functional similarities; however, basophils are true leukocytes
because they mature in the bone marrow and circulate in the
blood as mature cells with granules, whereas mast cell precur-
gastrointestinal, and genitourinary tracts. Survival time of sors (mononuclear cells without visible granules) leave the
eosinophils in humans is not well understood. In rats, tissue bone marrow and use the blood as a transit system to gain
survival time is about 6 days.23 access to the tissues where they mature and become granular.
Basophils are discussed first.
Eosinophil Functions Basophils are the least numerous of the circulating WBCs,
Eosinophils have multiple functions. Eosinophil granules are making up between 0.5% and 1.5% of circulating leukocytes.
full of a very large inventory of previously synthesized proteins,
including cytokines, chemokines, growth factors, and cationic Basophil Development
proteins. Basophil development is similar to eosinophil development in
There is more than one way for eosinophils to degranulate. that it requires the cytokines IL-3, IL-5, and GM-CSF; however,
By classical exocytosis, granules move to the plasma mem- transforming growth factor suppresses eosinophil differentia-
brane, fuse with the plasma membrane, and empty their con- tion and enhances basophil differentiation.31,32
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 143

Figure 12-11 Immature basophil (arrow). Note that the background cyto-
plasm is deeply basophilic with few large basophilic granules and there
appears to be a nucleolus.

Due to their small numbers, maturing basophils will not


be subdivided into separate morphologic phases. Rather,
immature and mature basophils will be described.
Immature basophils have round to somewhat lobulated B
nuclei with only slightly condensed chromatin. Nucleoli may
Figure 12-12 A, Mature basophil. Note that granules tend to obscure the
or may not be apparent. The cytoplasm is blue and contains nucleus and the background cytoplasm is only slightly basophilic. B, Electron
large blue-black secondary granules that are metachromatic micrograph of a basophil. (B from Carr JH, Rodak BF: Clinical hematology
(stain purple with toluidine blue) (Figure 12-11). Primary atlas, ed 3, Philadelphia, 2009, Saunders.)
azure granules may or may not be seen. Basophil granules are
water soluble and therefore may be dissolved if the blood film
is washed too much during the staining process. been regarded as the poor relatives of mast cells and minor
Mature basophils contain a lobulated nucleus that is often players in allergic inflammation because, like mast cells, they
difficult to see properly because of the granules lying on top. have immunoglobulin E (IgE) receptors on their surface mem-
The chromatin pattern, if visible, is clumped. Actual nuclear branes that, when cross-linked by antigen, result in granule
segmentation with visible filaments occurs rarely. The cyto- release.35 Today, something of a reawakening has occurred
plasm is colorless and contains large numbers of the character- regarding basophils and their functions in both innate and
istic metachromatic granules. If any granules have been adaptive immunity. Basophils are capable of releasing large
dissolved during the staining process, they often leave a quantities of subtype 2 helper T cell (TH2) cytokines such as
reddish purple rim surrounding what appears to be a vacuole IL-4 and IL-13 that regulate the TH2 immune response.36,37
(Figure 12-12). Basophils also induce B cells to synthesize IgE.38 Whereas mast
cells are the effectors of IgE-mediated chronic allergic inflam-
Basophil Kinetics mation, basophils function as initiators of the allergic inflam-
Basophil kinetics is poorly understood because of their very mation through the release of preformed cytokines.34 Basophil
small numbers. According to an early study, the time for bone activation is not restricted to antigen-specific IgE cross-linking,
marrow development and storage of basophils is 4.3 days 11 but can be prompted in nonsensitized individuals by a growing
hours, and their mean transit time in the peripheral blood is list of parasitic antigens, lectins, and viral superantigens,
3.7 days 21 hours.22 The life span of basophils is significantly binding to nonspecific IgE antibodies.39
longer than that of the other granulocytes. This has been attrib- The contents of basophil granules are not well known.
uted to the fact that, when they are activated by the cytokine Box 12-4 provides a short list of some of the substances released
IL-3, antiapoptotic pathways are initiated which cause the pro- by activated basophils. Moreover, mature basophils are evi-
longation of the basophil life span.33 dently capable of synthesizing granule proteins based on
activation signals. For example, basophils can be induced to
Basophil Functions produce a mediator of allergic inflammation known as gran-
Basophil functions are also poorly understood because of the zyme B.40 Mast cells can induce basophils to produce and
small numbers of these cells and the lack of animal models release retinoic acid, a regulator of immune and resident cells
such as basophil-deficient animals. Recently, however, an anti- in allergic diseases.41 Basophils may also play a role in angio-
basophil antibody has been developed and has served as a tool genesis through the expression of several forms of vascular
for studying basophil functions.34 In the past, basophils have endothelial growth factor and their receptors.42
144 PART II Hematopoiesis

BOX 12-4 Basophil Granules

Secondary Granules
Histamine
Platelet activating factor
Leukotriene C4
Interleukin-4
Interleukin-13
Vascular endothelial growth factor A
Vascular endothelial growth factor B
Chondroitin sulfates (e.g., heparin)

Finally, along with eosinophils, basophils are involved in


the control of helminth infections. They promote eosinophilia,
are associated with the differentiation of alternatively activated
macrophages in the lung, and contribute to efficient worm Figure 12-13 Tissue mast cell in bone marrow. Note that the nucleus is
expulsion.43 rounded and the cell is packed with large basophilic granules. Mast cells
tend to be a little larger than basophils (12 to 25m). These cells generally
Mast Cells make up only 1% of nucleated marrow cells. (From Carr JH, Rodak BF:
Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
Mast cells are not leukocytes. They are tissue cells. A brief
description of their development and function is included here
for two reasons: (1) their precursors circulate in the peripheral Monocyte Development
blood for a brief period on their way to their tissue destina- Monocyte development is similar to neutrophil development,
tions, and (2) mast cells have several phenotypic and func- because the two cell types share the GMP. Macrophage colony-
tional similarities with both basophils and eosinophils.44 stimulating factor is the major cytokine responsible for the
Although it has been well established that mast cell progeni- growth and differentiation of monocytes. The morphologic
tors (MCP) originate in the bone marrow, the exact pathway stages of monocyte development are monoblasts, promono-
of development has not been totally clarified. The bulk of cytes, and monocytes. Monoblasts in normal bone marrow are
evidence seems to indicate that the MCP arises from the so similar to myeloblasts morphologically that it is not produc-
GMP.45,46 Regardless of the lineage pathway, the major cytokine tive to attempt to separate the two. Malignant monoblasts in
responsible for mast cell maturation and differentiation is stem acute monoblastic leukemia are described in Chapter 36. There-
cell factor, also known as kit ligand.47 Once the MCP reaches fore, only promonocytes and monocytes are described here.
its tissue destination, complete maturation into mature mast Promonocytes are 12 to 18m in diameter, and their nucleus
cells occurs under the control of the local microenvironment is large in relation to their size (N:C ratio higher than 1). The
(Figure 12-13).48 There are two types of mast cells based on the nucleus may be round or, more frequently, slightly indented
presence or absence of chymase (C) along with tryptase (T) in or folded. The chromatin pattern is delicate, and at least
their granules (MCTC and MCT).49 one nucleolus should be apparent. The cytoplasm is blue and
Mast cells function as effector cells in allergic reactions contains scattered azure granules that are fewer and smaller
through the release of a wide variety of lipid mediators, prote- than those seen in promyelocytes (Figure 12-14). Electron
ases, proteoglycans, and cytokines as a result of cross-linking microscopic and cytochemical studies have shown that mono-
of IgE on the mast cell surface by specific allergens. Mast cells cyte azure granules are heterogeneous with regard to their
can also be activated independently of IgE, which leads to content of lysosomal enzymes, peroxidase, nonspecific ester-
inflammatory reactions. Mast cells can function as antigen- ases, and lysozyme.53
presenting cells to induce the differentiation of TH2 cells50; Monocytes appear to be larger than neutrophils (diameter of
therefore, mast cells act in both innate and adaptive immu- 15 to 20m) because they tend to stick to and spread out on
nity.51 In addition, mast cells can have antiinflammatory and glass or plastic. Their volume when assessed in solution by flow
immunosuppressive functions, and thus they can both enhance cytometry, however, is actually somewhat smaller than that of
and suppress features of the immune response.52 neutrophils. Monocytes are slightly immature cells whose ulti-
mate goal is to enter the tissues and mature into macrophages,
osteoclasts, or dendritic cells.
MONONUCLEAR CELLS
Their N:C ratio is 1. The nucleus may be round or oval but
Monocytes more frequently is deeply indented (horseshoe shaped) or
Monocytes make up between 2% and 10% of circulating leu- folded on itself. The chromatin pattern is looser than in the
kocytes, with between 96 and 1100 monocytes per microliter other leukocytes and has sometimes been described as lacelike
of whole blood. or stringy. This chromatin pattern underscores the slight
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 145

Figure 12-14 Large cell in the middle is a promonocyte. Note that the
nucleus is deeply indented and should not be confused with a neutrophil
band form (compare the chromatin patterns of the two). The cytoplasm is
deeply basophilic with azure granules that are much smaller than those seen
in progranulocytes. The azure granules in this cell are hard to see and give
the cytoplasm a slightly grainy appearance.

immaturity of monocytes. Nucleoli are generally not seen with


the light microscope; however, electron microscopy reveals
nucleoli in roughly half of circulating monocytes. Their cyto-
plasm is blue-gray with fine azure granules often referred to as
azure dust or a ground-glass appearance. Small cytoplasmic B
pseudopods or blebs may be seen. Cytoplasmic and nuclear Figure 12-15 A, Typical monocyte showing vacuolated cytoplasm, a
vacuoles may also be present (Figure 12-15). Based on flow contorted nucleus that tends to fold on itself, and very fine azure granules
cytometry, there are at least two subsets of monocytes: those that are sometimes referred to as azure dust. B, Electron micrograph of a
that are CD16+ and those that are CD16.54 monocyte. Note that the villi on the surface are much greater in number than
is seen on neutrophils. (B from Carr JH, Rodak BF: Clinical hematology atlas,
Monocyte/Macrophage Kinetics ed 3, Philadelphia, 2009, Saunders.)
The promonocyte pool consists of approximately 6 108 cells/
kg and they produce 7 106 monocytes/kg per hour. Under
normal circumstances, promonocytes undergo two mitotic divi- BOX 12-5 Monocyte Destinations
sions in 60 hours to produce a total of four monocytes. Under Differentiation into macrophages:
conditions of increased demand for monocytes, promonocytes In areas of inflammation or infection (inflammatory macrophages)
undergo four divisions to yield a total of 16 monocytes in 60 As resident macrophages in:
hours. There is no storage pool of monocytes in the bone Liver (Kupffer cells)
marrow.55 This is the reason why, when bone marrow recovers Lungs (alveolar macrophages)
from marrow failure, monocytes are seen in the peripheral Brain (microglia)
blood before neutrophils and a relative monocytosis may occur. Skin (Langerhans cells)
There is recent evidence, however, that a relatively large reservoir Spleen (splenic macrophages)
of immature monocytes resides in the subcapsular red pulp of Intestines (intestinal macrophages)
the spleen. Monocytes in this splenic reservoir appear to Peritoneum (peritoneal macrophages)
respond to tissue injury such as myocardial infarction by migrat- Bone (osteoclasts)
ing to the site of tissue injury to participate in wound healing.56 Synovial macrophages (type A cell)
Like neutrophils, monocytes in the peripheral blood can be Kidneys (renal macrophages)
found in a marginal pool and a circulating pool. Unlike with Reproductive organ macrophages
neutrophils, the marginal pool of monocytes is 3.5 times the Lymph nodes (dendritic cells)
circulating pool.57 Monocytes remain in the circulation approx-
imately 30 hours.58 Monocytes with different patterns of che-
mokine receptors have different target tissues and different tissues. Macrophages can be as large as 40 to 50m in diameter.
functions. Box 12-5 contains a list of the various tissue They usually have an oval nucleus with at least one rather
destinations of monocytes.59 Once in the tissues, monocytes prominent nucleolus and netlike (reticulated) chromatin
differentiate into macrophages, osteoclasts (Figure 12-16), or pattern. Their cytoplasm is pale, frequently vacuolated, and
dendritic cells depending on the microenvironment of the local often filled with debris of phagocytized cells or organisms.
146 PART II Hematopoiesis

surfaces (antigen-presenting cells). Because of this, they


interact with and activate both T lymphocytes and B
lymphocytes to initiate the adaptive immune response.
Dendritic cells are the most efficient and potent of the
antigen-presenting cells.
Housekeeping functions: These include (1) removal of debris
and dead cells at sites of infection or tissue damage, (2)
destruction of senescent red cells and maintenance of a
storage pool of iron for erythropoiesis, and (3) synthesis of
a wide variety of proteins, including complement compo-
nents, coagulation factors, prostaglandins, leukotrienes,
growth factors, and binding proteins such as transferrin.61
A
Lymphocytes
Lymphocytes are divided into three major groups: T cells, B
cells, and natural killer (NK) cells. T and B cells are major players
in adaptive immunity. NK cells make up a small percentage of
lymphocytes and are part of innate immunity. Adaptive immu-
nity has three characteristics: (1) It relies on an enormous
number of distinct lymphocytes, each having surface receptors
for a different specific molecular structure on a foreign antigen.
(2) After an encounter with a particular antigen, memory cells
are produced that will react faster and more vigorously to that
same antigen upon reexposure. (3) Self-antigens are ignored
under normal circumstances (referred to as tolerance).
Lymphocytes can be subdivided into two major categories:
those that participate in humoral immunity by producing
antibodies, and those that participate in cellular immunity
B by attacking foreign organisms or cells directly. Antibody-
producing lymphocytes are called B lymphocytes or simply B
Figure 12-16 A, Active marrow macrophage. B, Osteoclast. Both of
cells because they develop in the bone marrow. Cellular immu-
these cells are derived from monocytes.
nity is accomplished by two types of lymphocytes: T cells, so
named because they develop in the thymus, and NK cells,
The life span of macrophages in the tissues depends on which develop in both the bone marrow and the thymus.62,63
whether they are responding to inflammation or infection, or Lymphocytes are different from the other leukocytes in
are resident macrophages such as Kupffer cells or alveolar several ways, including the following:
macrophages. Resident macrophages survive far longer than 1. Lymphocytes are not end cells. They are resting blast forms
tissue neutrophils. For example, Kupffer cells have a life span capable, when stimulated, of transforming into active
of approximately 21 days.60 Inflammatory macrophages, on blasts that undergo mitosis to produce both memory and
the other hand, have a life span measured in hours. effector cells.
2. Unlike other leukocytes, lymphocytes recirculate from the
Monocyte/Macrophage Functions blood to the tissues and back to the blood.
Functions of monocytes/macrophages are numerous and 3. B and T lymphocytes are capable of recombining gene
varied. They can be subdivided into innate immunity, adaptive segments to produce a wide variety of surface receptors
immunity, and housekeeping functions. and antibodies.
Innate immunity: Monocytes/macrophages recognize a wide 4. Although early lymphocyte progenitors such as the common
range of bacterial pathogens by means of pattern recogni- lymphocyte progenitor originate in the bone marrow, T and
tion receptors (toll-like receptors) that stimulate inflamma- NK lymphocytes usually develop outside of the bone marrow.
tory cytokine production and phagocytosis. Macrophages For these reasons, lymphocyte kinetics is extremely compli-
can synthesize nitric oxide, which is cytotoxic against viruses, cated, not well understood, and beyond the scope of this chapter.
bacteria, fungi, protozoa, helminths, and tumor cells.17 Lymphocytes make up between 20% and 40% of circulating
Monocytes and macrophages also have Fc receptors and leukocytes with 960 to 4400 lymphocytes per microliter of
complement receptors. Hence, they can phagocytize foreign whole blood.
organisms or material that has been coated with antibodies
or complement. Lymphocyte Development
Adaptive immunity: Both macrophages and dendritic cells For both B and T cells development can be subdivided
degrade antigen and present antigen fragments on their into antigen-independent and antigen-dependent phases.
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 147

Antigen-independent lymphocyte development occurs in the


bone marrow and thymus (sometimes referred to as central or
primary lymphatic organs), whereas antigen-dependent lympho-
cyte development occurs in the spleen, lymph nodes, tonsils,
and nonencapsulated aggregates of lymphocytes such as the
Peyer patches in the intestinal wall (sometimes referred to as
peripheral or secondary lymphatic organs).
B lymphocytes develop initially in the bone marrow and go
through three stages known as pro-B, pre-B, and immature B
cells. It is during these stages that gene rearrangement occurs to
produce unique immunoglobulin chains. The morphology of
these primitive B cells has not been established. Immature B
cells (sometimes referred to as naive B cells) leave the bone A
marrow to migrate to secondary lymphatic organs, where they
take up residence in specific zones such as lymph node folli-
cles. The morphology of immature B cells (also known as
hematogones) is similar to that of the cells seen in acute
lymphocytic leukemia.64 They are cells with a homogeneous
nuclear chromatin pattern and extremely scanty cytoplasm
(Figure 12-17). These cells are normally seen in newborn
peripheral blood and bone marrow.
It is in the secondary lymphatic organs or in the blood
where B cells may come in contact with antigen, which results
in cell division (blastogenesis) and the production of memory
cells as well as effector cells. Effector B cells are antibody-
producing cells known as plasma cells and plasmacytoid lympho-
cytes (Figure 12-18). B
Approximately 10% to 15% of circulating lymphocytes are Figure 12-18 Effector cells of the B lymphocyte line. A, Plasma cell.
B cells. Resting B lymphocytes cannot be distinguished mor- B, Plasmacytoid lymphocyte.
phologically from resting T lymphocytes. Resting lymphocytes
are small (around 9m in diameter) and the N:C ratio is 1:1.
The chromatin is arranged in blocks and the nucleolus is rarely
seen, although it is present (Figure 12-19).
T lymphocytes develop initially in the thymusa lymphoepi-
thelial organ located in the upper mediastinum.65 Lymphoid
progenitor cells migrate from the bone marrow to the thymic
cortex where, under the regulation of cytokines produced by
thymic epithelial cells, they progress through stages known as
pro-T, pre-T, and immature T cells. During these phases they

B
Figure 12-17 Dark cell to the right of center is an immature B lymphocyte Figure 12-19 A, Small resting lymphocyte. B, Electron micrograph of
(also known as a hematogone). Note the extremely scanty cytoplasm. This a small lymphocyte. (B from Carr JH, Rodak BF: Clinical hematology atlas,
was taken from the bone marrow of a newborn infant. ed 3, Philadelphia, 2009, Saunders.)
148 PART II Hematopoiesis

Figure 12-20 Three cells representing blastogenesis. A small resting


lymphocyte (A) is stimulated by antigen and begins to enlarge to form a Figure 12-21 A large granular lymphocyte that could be either a cytotoxic
medium to large lymphocyte (B). The nucleus reverts from a clumped to a T lymphocyte or a natural killer lymphocyte.
delicate chromatin pattern with nucleoli, and the cell becomes a blast that
is capable of dividing to form effector cells or memory cells (C). azurophilic granules that are peroxidase negative. They have
been referred to frequently as large granular lymphocytes (Figure
12-21).67 Approximately 2% of circulating leukocytes are NK
undergo gene rearrangement to produce T-cell receptors that lymphocytes.
are unique to each T cell. T cells whose receptors react with
self-antigens are allowed to die through neglect.66 In addition, Lymphocyte Functions
T cells are subdivided into two major categories depending on Functions can be addressed according to the various types of
whether or not they have CD4 or CD8 antigen on their surfaces. lymphocyte.
Immature T cells then proceed to the thymic medulla, where B lymphocytes are essential for antibody production. In addi-
further removal and destruction of self-reactive T cells occurs. tion, they have a role in antigen presentation to T cells and
The remaining immature T cells (or naive T cells) then leave the may be necessary for optimal CD4 activation. B cells also
thymus and migrate to secondary lymphatic organs, where they produce cytokines that regulate a variety of T-cell and antigen-
take up residence in specific zones such as the paracortical areas. presenting cell functions.68
Roughly 85% of circulating lymphocytes are T cells. T lymphocytes can be divided into CD4+ T cells and CD8+
T cells in secondary lymphatic organs or in the circulating T cells.
blood eventually come in contact with antigen. This results in CD4+ effector lymphocytes are further subdivided into TH1,
cell activation, blastogenesis, and the production of either TH2, TH17, and Treg (CD4+CD25+ regulatory T cells) cells. TH1
memory cells or effector T cells or both (Figure 12-20). The trans- cells mediate immune responses against intracellular patho-
formation of resting lymphocytes into blast forms is the source gens. TH2 cells mediate host defense against extracellular para-
of so-called medium and large lymphocytes that have increased sites, including helminths. They are also important in the
amounts of cytoplasm and usually make up only about 10% of induction of asthma and other allergic diseases. TH17 cells
circulating lymphocytes. The morphology of effector T cells are involved in the immune responses against extracellular
varies with the subtype of T cell involved, and they are often bacteria and fungi. Treg cells play a role in maintaining self-
referred to as reactive or variant lymphocytes (see Chapter 28). tolerance by regulating immune responses.69
NK cells may develop in either the bone marrow or CD8+ effector lymphocytes are capable of killing target cells
the thymus. They are a heterogeneous group of cells by secreting granules containing granzyme and perforin or by
with respect to their surface antigens. The majority are activating apoptotic pathways in the target cell.70 These cells
CD56+CD16+CD3CD8. Morphologic stages of developing are sometimes referred to as cytotoxic T lymphocytes.
NK cells have not been described. The mature NK cell is rela- NK lymphocytes function as part of innate immunity in that
tively large compared with other resting lymphocytes because they are capable of killing certain tumor cells and virus-infected
of an increased amount of cytoplasm. Its cytoplasm contains cells without prior sensitization.

SUMMARY
In the adult human, the bone marrow is the source of all leuko- Granulocytes are classified according to their staining character-
cytes as well as certain tissue cells such as mast cells, osteo- istics and the shape of their nuclei. Neutrophils are a major
clasts, macrophages, dendritic cells, and endothelial cells. Some component of innate immunity as phagocytes, eosinophils are
leukocyte progenitors leave the bone marrow to develop in the involved in allergic reactions and helminth destruction, and
thymus (T cells, NK cells, and dendritic cells). basophils function as initiators of allergic reactions as well as
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 149

helminth destruction. All three granulocytes also have other impor- Monocyte development can be subdivided into the promonocyte,
tant functions. monocyte, and macrophage stages, each with specific morpho-
Neutrophil development can be subdivided into specific stages, logic characteristics.
with cells at each stage having specific morphologic characteris- The majority of lymphocytes are involved in adaptive immunity.
tics (myeloblast, promyelocyte, myelocyte, metamyelocyte, band B lymphocytes produce antibodies against foreign organisms
form, and segmented form). Various granule types are produced or cells, and T lymphocytes mediate the immune response
during neutrophil development, each with specific contents. against intracellular and extracellular invaders. Both B and T
Eosinophil development can also be subdivided into specific lymphocytes produce memory cells for specific antigens so
stages, although eosinophilic myeloblasts are not recognizable that the immune response is quicker if the same antigen is
and eosinophil promyelocytes are extremely rare. encountered again.
Basophil development is difficult to describe, and basophils have Lymphocyte development is complex, and morphologic divisions
been divided simply into immature and mature basophils. are not practical because a large number of lymphocytes develop
Mononuclear cells consist of monocytes and lymphocytes. Mono- in the thymus. Benign B-lymphocyte precursors (hematogones) as
cytes are precursors to tissue cells such as osteoclasts, macro- well as B-lymphocyte effector cells (plasma cells and plasmacy-
phages, and dendritic cells. As a group, they perform several toid lymphocytes) have been described. NK lymphocytes and cyto-
functions as phagocytes. toxic T cells also have a distinct and similar morphology.

R E V I E W Q UESTIONS
1. Neutrophils and monocytes are direct descendants of a 6. Which of the following cell types is capable of differ
common progenitor known as: entiating into osteoclasts, macrophages, or dendritic
a. CMP cells?
b. GMP a. Neutrophils
c. CFU-GEMM b. Lymphocytes
d. HSC c. Monocytes
d. Eosinophils
2. Neutrophils contain four different types of granules. The
granules containing adhesive molecules such as CD11b/ 7. Macrophages aid in adaptive immunity by:
CD18 that facilitate migration and diapedesis are referred a. Synthesizing complement components
to as: b. Degrading antigen and presenting it to lymphocytes
a. Primary c. Storing iron from senescent red cells
b. Secondary d. Ingesting and digesting organisms that neutrophils
c. Tertiary cannot
d. Secretory
8. Which of the following is the final stage of B-cell matura-
3. Some eosinophil granules contain a core crystalline struc- tion after activation by antigen?
ture composed mostly of: a. Large, granular lymphocyte
a. Major basic protein b. Reactive lymphocyte
b. Lysosomal enzymes c. Plasma cell
c. Peroxidase d. Immunoblast
d. Neurotoxin
9. A large number of T lymphocytes die or are destroyed in
4. Basophils and mast cells have high-affinity surface recep- the thymus because they:
tors for which immunoglobulin? a. React to self-antigens
a. A b. Do not have proper surface markers
b. D c. Lack appropriate morphology
c. E d. Experience gene rearrangements that lead to
d. G apoptosis

5. Basophil granules may not survive the staining process 10. The following is unique to both B and T lymphocytes and
very well because they are: occurs during their early development:
a. Highly acidic a. Synthesis of surface antigens CD4 and CD8
b. Soluble in water b. Blastogenesis
c. Crystalline c. Synthesis of immunoglobulins
d. Soluble in alcohol d. Gene rearrangement
150 PART II Hematopoiesis

REFERENCES
1. von Vietinghoff S, Ley K: Homeostatic regulation of blood recruitment upon MHC class Irestricted thymocyte deletion.
neutrophil counts. J Immunol 181:5183-5188, 2008. J Immunol 165:1965-1975, 2000.
2. Price TH, Chatta GS, Dale DC: Effect of recombinant granu- 26. Hogan SP, Rosenberg HF, Moqbel R, et al: Eosinophils: bio-
locyte colony-stimulating factor on neutrophil kinetics in logical properties and role in health and disease. Clin Exp
normal young and elderly humans. Blood 88:335-340, 1996. Allergy 38:709-750, 2008.
3. Terstappen LW, Huang S, Safford M, et al: Sequential 27. Spencer LA, Szela CT, Perez SA, et al: Human eosinophils
generations of hematopoietic colonies derived from single constitutively express multiple Th1, Th2, and immunoregula-
nonlineage-committed CD34+CD38 progenitor cells. Blood tory cytokines that are secreted rapidly and differentially.
77:1218-1227, 1991. J Leukoc Biol 85:117-123, 2009.
4. Manz MG, Miyamoto T, Akashi K, et al: Prospective isolation 28. Hagan P, Wilkins HA, Blumenthal UJ, et al: Eosinophilia
of human clonogenic common myeloid progenitors. Proc and resistance to Schistosoma haematobium in man. Parasite
Natl Acad Sci U S A 99:11872-11877, 2002. Immunol 7:625-632, 1985.
5. Giebel B, Punzel M: Lineage development of hematopoietic 29. Phipps S, Benyahia F, Ou TT, et al: Acute allergen-induced
stem and progenitor cells. Biol Chem 389:813-824, 2008. airway remodeling in atopic asthma. Am J Respir Cell Mol Biol
6. Iwasaki H, Akashi K: Hematopoietic developmental path- 31:626-632, 2004.
ways: on cellular basis. Oncogene 26: 6687-6696, 2007. 30. Walsh RE, Gaginella TS: The eosinophil in inflammatory
7. Faurschou M, Borregaard N: Neutrophil granules and secre- bowel disease. Scand J Gastroenterol 26:1217-1224, 1991.
tory vesicles in inflammation. Microbes Infect 5:1317-1327, 31. Sillaber C, Geissler K, Scherrer R, et al: Type beta transforming
2003. growth factors promote interleukin-3 (IL-3)dependent dif-
8. Cornbleet PJ: Clinical utility of the band count. Clin Lab Med ferentiation of human basophils but inhibit IL-3dependent
22:101-136, 2002. differentiation of human eosinophils. Blood 80:634-641,
9. Dancey JT, Deubelbeiss KA, Harker LA, et al: Neutrophil 1992.
kinetics in man. J Clin Invest 58:705-715, 1976. 32. Denburg JA: Basophil and mast cell lineages in vitro and in
10. Cartwright GE, Athens JW, Wintrobe MM: The kinetics of vivo. Blood 79:846-860, 1992.
granulopoiesis in normal man. Blood 24:780-803, 1964. 33. Didichenko SA, Spiegl N, Brunner T, et al: IL-3 induces a
11. Cowburn AS, Condliffe AM, Farahi N, et al: Advances in Pim1-dependent antiapoptotic pathway in primary human
neutrophil biology: clinical implications. Chest 134:606-612, basophils. Blood 112:3949-3958, 2008.
2008. 34. Obata K, Mukai K, Tsujimura Y, et al: Basophils are essential
12. Hetherington SV, Quie PG: Human polymorphonuclear leu- initiators of a novel type of chronic allergic inflammation.
kocytes of the bone marrow, circulation and marginated Blood 110:913-920, 2007.
pool: function and granule protein content. Am J Hematol 35. Sullivan BM, Locksley RM: Basophils: a nonredundant con-
20:235-246, 1985. tributor to host immunity. Immunity 30:12-20, 2009.
13. Ley K, Laudanna C, Cybulsky MI, et al: Getting to the site of 36. Schroeder JT, MacGlashan DW Jr, Lichtenstein LM: Human
inflammation: the leukocyte adhesion cascade updated. Nat basophils: mediator release and cytokine production. Adv
Rev Immunol 7:678-689, 2007. Immunol 77:93-122, 2001.
14. Mayadas TN, Cullere X: Neutrophil 2 integrins: moderators 37. Gibbs BF: Human basophils as effectors and immunomodu-
of life or death decisions. Trends Immunol 26:388-395, 2005. lators of allergic inflammation and innate immunity. Clin Exp
15. Burg ND, Pillinger MH: The neutrophil: function and Med 5:43-49, 2005.
regulation in innate and humoral immunity. Clin Immunol 38. Gauchat JF, Henchoz S, Mazzei G, et al: Induction of human
99:7-17, 2001. IgE synthesis in B cells by mast cells and basophils. Nature
16. Stuart LM, Ezekowitz RA: Phagocytosis: elegant complexity. 365:340-343, 1993.
Immunity 22:539-550, 2005. 39. Falcone FH, Zillikens D, Gibbs BF: The 21st century renais-
17. Dale DC, Boxer L, Liles WC: The phagocytes: neutrophils and sance of the basophil? Current insights into its role in allergic
monocytes. Blood 112:935-945, 2008. responses and innate immunity. Exp Dermatol 15:855-864,
18. Brinkmann V, Zychlinsky A: Beneficial suicide: why neutro- 2006.
phils die to make NETs. Nat Rev Microbiol 5:577-582, 2007. 40. Tschopp CM, Spiegl N, Didichenko S, et al: Granzyme B,
19. Boyce JA, Friend D, Matsumoto R, et al: Differentiation in a novel mediator of allergic inflammation: its induction
vitro of hybrid eosinophil/basophil granulocytes: autocrine and release in blood basophils and human asthma. Blood
function of an eosinophil developmental intermediate. J Exp 108:2290-2299, 2006.
Med 182:49-57, 1995. 41. Spiegl N, Didichenko S, McCaffery P, et al: Human basophils
20. Melo RCN, Spencer LA, Perez SAC, et al: Vesicle-mediated activated by mast cell-derived IL-3 express retinaldehyde
secretion of human eosinophil granule-derived major basic dehydrogenase-11 and produce the immunoregulatory medi-
protein. Lab Invest 89:769-781, 2009. ator retinoic acid. Blood 112:3762-3771, 2008.
21. Giembycz MA, Lindsay MA: Pharmacology of the eosinophil. 42. de Paulis A, Preverte N, Fiorentino I, et al: Expression and
Pharmacol Rev 51:213-339, 1999. functions of the vascular endothelial growth factors and their
22. Steinbach KH, Schick P, Trepel F, et al: Estimation of kinetic receptors in human basophils. J Immunol 177:7322-7331,
parameters of neutrophilic, eosinophilic and basophilic gran- 2006.
ulocytes in human blood. Blut 39:27-38, 1979. 43. Ohnmacht C, Voehringer D: Basophil effector function and
23. Foot EC: Eosinophil turnover in the normal rat. Br J Haematol homeostasis during helminth infections. Blood 113:2816-
11:439-445, 1965. 2825, 2009.
24. Melo RC, Spencer LA, Dvorak AM, et al: Mechanisms of 44. Valent P: The phenotype of human eosinophils, basophils
eosinophil secretion: large vesiculotubular carriers mediate and mast cells. J Allergy Clin Immunol 94:1177-1183, 1994.
transport and release of granule-derived cytokines and other 45. Kirshenbaum AS, Goff JP, Semere T, et al: Demonstration that
proteins. J Leukoc Biol 83:229-236, 2008. human mast cells arise from a progenitor cell population that
25. Throsby M, Herbelin A, Pleau JM, et al: CD11c+ eosinophils is CD34+, c-kit+, and expresses aminopeptidase N (CD13).
in the murine thymus: developmental regulation and Blood 94:2333-2342, 1999.
CHAPTER 12 Leukocyte Development, Kinetics, and Functions 151
46. Gurish MF, Boyce JA: Mast cells: ontogeny, homing, and 58. van Furth R, Cohn ZA: The origin and kinetics of mononu-
recruitment of a unique innate effector cell. J Allergy Clin clear phagocytes. J Exp Med 128:415-435, 1968.
Immunol 117:1285-1291, 2006. 59. Kumar S, Jack R: Origin of monocytes and their differentia-
47. Valent P, Spanblchl E, Sperr WR, et al: Induction of differ- tion to macrophages and dendritic cells. J Endotox Res 12:278-
entiation of human mast cells from bone marrow and 284, 2006.
peripheral blood mononuclear cells by recombinant human 60. Crofton RW, Diesselhoff-den Dulk MM, van Furth R: The
stem cell factor/kit-ligand in long term culture. Blood 80:2237- origin and kinetics of the Kupffer cells in the normal steady
2245, 1992. state. J Exp Med 148:1-17, 1978.
48. Kambe, N, Hiramatsu H, Shimonaka M, et al: Development 61. Taylor GA, Weinberg JB: Mononuclear phagocytes. In Green
of both human connective tissue-type and mucosal-type mast JP, Foerster J, Rodgers GM, et al, editors: Wintrobes clinical
cells in mice from hematopoietic stem cells with identical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
distribution pattern to human body. Blood 103:860-867, Health/Lippincott Williams & Wilkins, pp 249-280.
2004. 62. Lotzov E, Savary CA, Champlin RE: Genesis of human onco-
49. Irani AA, Schechter NM, Craig SS, et al: Two types of human lytic natural killer cells from primitive CD34+CD33 bone
mast cells that have distinct neutral protease compositions. marrow progenitors. J Immunol 150:5263-5269, 1993.
Proc Natl Acad Sci U S A 83:4464-4468, 1986. 63. Res P, Martnez-Cceres E, Cristina Jaleco A, et al:
50. Nakano N, Nishiyama C, Yagita H, et al: Notch signaling CD34+cd38dim cells in the human thymus can differentiate
confers antigen-presenting cell functions on mast cells. into T, natural killer, and dendritic cells but are distinct from
J Allergy Clin Immunol 123:74-81, 2009. pluripotent stem cells. Blood 87:5196-5206, 1996.
51. Heib V, Becker M, Taube C, et al: Advances in the understand- 64. Intermesoli T, Mangili G, Salvi A, et al: Abnormally expanded
ing of mast cell functions. Br J Haematol 142:683-694, 2008. pro-B hematogones associated with congenital cytomegalo-
52. Galli SJ, Grimbaldeston M, Tsai M: Immunomodulatory mast virus infection. Am J Hematol 82:934-936, 2007.
cells: negative, as well as positive, regulators of immunity. 65. Haynes BF: The human thymic microenvironment. Adv
Nat Rev Immunol 8:478-486, 2008. Immunol 36:87-142, 1984.
53. Nichols BA, Bainton DF, Farquhar MG: Differentiation of 66. von Boehmer H, Teh HS, Kisielow P: The thymus selects the
monocytesorigin, nature and fate of their azurophil gran- useful, neglects the useless and destroys the harmful. Immunol
ules. J Cell Biol 50:498-515, 1971. Today 10:57-61, 1989.
54. Geissmann F, Jung S, Littman DR: Blood monocytes consist 67. Grossi CE, Cadoni A, Zicca A, et al: Large granular lympho-
of two principal subsets with distinct migratory properties. cytes in human peripheral blood: ultrastructural and cyto-
Immunity 19:71-82, 2003. chemical characterization of the granules. Blood 59:277-283,
55. Meuret G, Bammert J, Hoffmann G: Kinetics of human 1982.
monocytopoiesis. Blood 44:801-816, 1974. 68. LeBien TW, Tedder TF: B lymphocytes: how they develop and
56. Swirski FK, Nahrendorf M, Etzrodt M, et al: Identification of function. Blood 112:1570-1580, 2008.
splenic reservoir monocytes and their deployment to inflam- 69. Zhu J, Paul WE: CD4 T cells: fates, functions and faults. Blood
matory sites. Science 325:612-616, 2009. 112:1557-1569, 2008.
57. Meuret G, Batara E, Frste HO: Monocytopoiesis in normal 70. Rufer N, Zippelius A, Batard P, et al: Ex vivo characterization
man: pool size, proliferation activity and DNA synthesis time of human CD8+ T subsets with distinct replicative history and
of promonocytes. Acta Haematol 54:261-270, 1975. partial effector functions. Blood 102:1779-1787, 2003.
13 Platelet Production, Structure,
and Function
George A. Fritsma

OUTLINE OBJECTIVES
Megakaryocytopoiesis After completion of this chapter, the reader will be able to:
Endomitosis
Megakaryocyte Progenitors 1. Diagram megakaryocytopoiesis, including mega- 4. Recount platelet function, including adhesion, aggre-
Terminal Megakaryocyte karyocyte localization, growth factor control, endomi- gation, and secretion.
Differentiation tosis, and platelet shedding. 5. Reproduce the biochemical pathways of platelet
Thrombocytopoiesis (Platelet
2. Describe the ultrastructure of resting platelets in the activation, including integrins, G proteins, the eico-
Shedding)
Hormones and Cytokines of circulation, including the plasma membrane, tubules, sanoid, and the diacylglycerol-inositol triphosphate
Megakaryocytopoiesis microfibrils, and granules. pathway.
Platelets 3. List the important platelet receptors and their ligands.
Platelet Ultrastructure
Resting Platelet Plasma
Membrane CASE STUDY
Surface-Connected
After studying this chapter, the reader should be able to respond to the following case study:
Canalicular System
Dense Tubular System
A 35-year-old woman noticed multiple pinpoint red spots and bruises on her arms and legs. The
Platelet Plasma Membrane
Receptors That Provide for hematologist confirmed the presence of petechiae, purpura, and ecchymoses on her extremities and
Adhesion ordered a complete blood count, prothrombin time, and partial thromboplastin time. The platelet
Platelet Plasma Membrane count was 35 109/L, the mean platelet volume was 13.2fL, and the platelets appeared large on
Receptors That Provide the Wright-stained peripheral blood film. Other complete blood count parameters and the coagula-
for Activation: The
tion parameters were within normal limits. A Wright-stained bone marrow aspirate revealed 10 to
Seven-Transmembrane
Receptors 12 small unlobulated megakaryocytes per low-power field.
Additional Platelet Membrane 1. What type of bleeding is indicated by these signs and symptoms?
Receptors 2. What is the probable cause of the bleeding?
Platelet Cytoskeleton: 3. Is the thrombocytopenia the result of inadequate bone marrow production?
Microfilaments and
4. List the growth factors involved in recruiting megakaryocyte progenitors.
Microtubules
Platelet Granules:
-Granules, -Granules
and Lysosomes
Platelet Activation
Adhesion: Platelets
Reversibly Bind Elements MEGAKARYOCYTOPOIESIS
of the Vascular Matrix
Aggregation: Platelets Platelets are anucleate blood cells that circulate in amounts of 150 to 400 109/L, with mean counts
Irreversibly Bind Each slightly higher in women than in men.1 Platelets trigger primary hemostasis on exposure to endothe-
Other
lial, subendothelial, and plasma procoagulants in blood vessel injury. On a Wright-stained wedge-
Secretion: Activated Platelets
Release Granular Contents preparation blood film, platelets are distributed throughout the red blood cell monolayer at 7 to 21
Platelet Activation Pathways per 100 field. They have an average diameter of 2.5m, corresponding to a mean platelet volume
G Proteins (MPV) of 8 to 10fL in an isotonic suspension, as determined using laboratory profiling instruments.2
Eicosanoid Synthesis Their internal structure, although complex, is granular but scarcely visible using light microscopy.
Inositol Triphosphate
Platelets arise from unique bone marrow cells called megakaryocytes. Megakaryocytes are among the
Diacylglycerol Activation
Pathway largest cells in the body and are polyploid, which means that they possess multiple chromosome
copies within a single cell. On a Wright-stained bone marrow aspirate film, each megakaryocyte is 30
to 50m in diameter with a multilobulated nucleus and abundant granular cytoplasm. In healthy

152
CHAPTER 13 Platelet Production, Structure, and Function 153

Abluminal Megakaryocyte
Nerve surface

Nutrient
artery

Central
vein

Venous
sinus Lumen of venous sinus

Hematopoietic Endothelial cells


tissue

Figure 13-1 Megakaryocytes localize to abluminal surface of sinusoid-lining endothelial cells.

intact bone marrow tissue, megakaryocytes cluster in the extra- and cannot be distinguished by Wright-stained light micros-
vascular compartment adjacent to the abluminal membrane copy. The BFU-Meg and CFU-Meg are diploid and participate
(the surface opposite the lumen) of venous sinusoid endothe- in normal mitosis, maintaining a pool of megakaryocyte pro-
lial cells (Figure 13-1).3 Myelocytic and erythrocytic precursor genitors. Their proliferative (mitotic) properties are reflected in
cells, which locate further from the endothelial cells, may cross their ability to form colonies of hundreds (BFUs) or scores
the megakaryocyte cytoplasm to reach the sinusoid lumen, a (CFUs) of progeny in culture (Figure 13-2). In contrast to the
faux phagocytosis known as emperopolesis.4 Megakaryocytes are BFU-Meg and CFU-Meg, the LD-CFU-Meg has little prolifera-
also harvested from the lungs.5 In a normal Wright-stained tive capacity and produces few cells, but begins the progress
bone marrow aspirate film, the microscopist may identify two through endomitosis to reach increased nuclear ploidy.
to four megakaryocytes per 10 low-power field. The LD-CFU-Meg may be a transitional, or promegakaryo-
blast, stage in which polyploidy is first established, but
Endomitosis its morphology is indistinguishable from that of small lym-
Megakaryocyte maturation is marked by a mysterious form of phocytes. At the LD-CFU-Meg point of development, mega-
mitosis that lacks telophase and cytokinesis (separation into karyocyte progenitors enter terminal differentiation, as they lose
daughter cells) and is called endomitosis. In endomitosis, DNA their ability to undergo normal mitosis but continue with
replication proceeds to the production of 8N, 16N, or 32N endomitosis.
ploidy with duplicated sets of chromosomes, but no cell divi- In specialty laboratories, immunologic probes and flow
sion.6 Some megakaryocyte nuclei replicate five times, reaching cytometry are employed to identify megakaryocyte progenitors.
128N; this level of ploidy is unusual, however, and may signal Useful flow cytometric progenitor markers are the general stem
hematologic disease. Megakaryocytes employ their copious cell marker CD34, HLA-DR, and platelet glycoprotein IIIa (GP
DNA to synthesize abundant cytoplasm, which differentiates IIb/IIIa, CD41).8 Platelet peroxidase, localized in the endoplas-
into platelets. A single megakaryocyte may shed 2000 to 4000 mic reticulum of progenitors and megakaryoblasts, also may
platelets. In an average-size healthy human there are 108 be identified by cytochemical stain in transmission electron
megakaryocytes producing 1011 platelets per day. The key to microscopy. Identical peroxidase activity is localized to the
endomitosis is the loss of spindle fiber orientation at the dense tubular system of mature platelets.9
point of telophase, so that chromosomes do not proceed from
equatorial plates to polar bodies, but rather duplicate in place. Terminal Megakaryocyte Differentiation
Cytokinesis is arrested, a cell-cycle adaptation found in no Megakaryocyte progenitors leave the proliferative phase and
other normal human cell. enter terminal differentiation, a series of stages in which
microscopists begin to recognize their unique Wright-stained
Megakaryocyte Progenitors morphology in bone marrow aspirate films (Figure 13-3) or
The megakaryocyte-erythrocyte progenitor (see Figure 7-13) dif- hematoxylin and eosinstained bone marrow biopsy sections
ferentiates into the megakaryocyte lineage under the influence (Table 13-1).
of the hormone thrombopoietin (TPO) and a series of cyto- Morphologists call the least differentiated megakaryocyte
kines (see Table 13-3). There are three megakaryocyte lineage- precursor the MK-I stage or megakaryoblast. Although they no
committed progenitor stages, defined by their culture colony longer look like small lymphocytes, megakaryoblasts cannot
characteristics. In order of differentiation, these are the least be reliably distinguished from myeloblasts or pronormoblasts
mature burst-forming unit (BFU-Meg), the colony-forming unit using light microscopy (Figure 13-4). The morphologist may
(CFU-Meg), and the most mature light-density CFU (LD-CFU- see plasma membrane blebs, blunt projections from the margin
Meg).7 All three progenitor stages resemble small lymphocytes that resemble platelets. At this stage the megakaryocyte begins
154 PART II Hematopoiesis

Megakaryocyte-
erythrocyte TPO
progenitor Meg-CSF
IL-3

CFU-GEMM

BFU-Meg
Mitosis

CFU-Meg

LD-CFU-Meg Endomitosis

Figure 13-2 Three stages of megakaryocyte progenitors. Burst-forming unitmegakaryocyte (BFU-Meg) clones hundreds of daughter cells, colony-forming
unitmegakaryocyte (CFU-Meg) clones scores of daughter cells, and low-density colony-forming unitmegakaryocyte (LD-CFU-Meg) undergoes the first stage
of endomitosis. IL-3, Interleukin-3; Meg-CSF, megakaryocyte colony-stimulating factor; TPO, thrombopoietin.

LD-CFU-Meg MK-I MK-II MK-III


TPO
1
IL-1

Proplatelet Platelet
process shedding

Figure 13-3 Morphologically identifiable megakaryocytes of the terminal megakaryocyte differentiation compartment. IL-11, Interleukin 11; LD-CFU-Meg,
low-density colony forming unitmegakaryocyte; MK-I, megakaryocyte I or megakaryoblast; MK-II, megakaryocyte II or promegakaryocyte; MK-III, megakaryo-
cyte III or megakaryocyte; TPO, thrombopoietin.

to develop most of its cytoplasmic ultrastructure, including


procoagulant-laden -granules, -granules (dense bodies), and
the demarcation system (DMS).10
The later section on mature platelet ultrastructure discusses
-granules and -granules with their contents. The DMS is a
series of membrane-lined channels that invade from the plasma
membrane and grow over the course of terminal differentiation
to subdivide the entire cytoplasm. The DMS is biologically
identical to the plasma membrane and ultimately delineates
the individual platelets during thrombocytopoiesis.
If there is a clinical need, immunologic probes or cyto-
chemical markers are employed for definitive identification by
flow cytometry (Table 13-2). In addition to the progenitor
markers listed previously, the megakaryoblast may be identi-
Figure 13-4 The megakaryoblast is similar to the myeloblast and pronor- fied using immunologic probes for the more differentiated
moblast, which makes identification by morphology alone inadvisable. membrane structures GP Ib, which is part of the GP Ib/IX/V
CHAPTER 13 Platelet Production, Structure, and Function 155

von Willebrand factor (VWF) adhesion receptor (CD42); mpl, 50-m diameter (Figures 13-5 and 13-6). The nucleus is
which is the TPO receptor site; or cytoplasmic VWF, detected intensely indented or lobulated, and the degree of lobulation
by histochemical immunostaining rather than flow cytometry. is imprecisely proportional to ploidy. (When necessary, ploidy
CD36, which is GP IV, becomes visible in the developing MK-II levels are measured using mepacrine, a nucleic acid dye, in
stage, and fibrinogen may be detected by immunostaining in megakaryocyte flow cytometry.11) The chromatin is variably
the fully developed megakaryocyte.
Nuclear lobularity first becomes apparent as indentation at
the 4N replication stage, rendering the cell identifiable as an
MK-II stage megakaryocyte by light microscopy. The morpho
logist seldom makes the effort to distinguish the MK-II and
MK-III stages during a routine examination of a bone marrow
aspirate film.
The megakaryocyte reaches its full ploidy level by the end
of the MK-II stage. At the MK-III stage, the megakaryocyte is
easily recognized at 10 magnification on the basis of its 30- to

TABLE 13-1 Features of the Three Terminal


Megakaryocyte Differentiation Stages
MK-I MK-II MK-III
Figure 13-5 Promegakaryocyte (MK-II). Cytoplasm is abundant, and the
% Precursors 20 25 55 nucleus shows minimal lobularity.
Diameter 14-18m 15-40m 30-50m
Nucleus Round Indented Multilobed
Nucleoli 2-6 Variable Not visible
Chromatin Homogeneous Condensed Deeply but variably
condensed
Nucleus-to- 3:1 1:2 1:4
cytoplasm
ratio
Mitosis Absent Absent Absent
Endomitosis Present Ends Absent
Cytoplasm Basophilic Basophilic and Eosinophilic and
granular granular
-granules Present Present Present
-granules Present Present Present
Demarcation Present Present Present
system Figure 13-6 Megakaryocyte. The single nucleus is markedly lobulated
with variable stained, basophilic chromatin. The cytoplasm is azurophilic and
MK-I, Megakaryoblast; MK-II, promegakaryocyte; MK-III, megakaryocyte. granular with evidence of the demarcation system.

TABLE 13-2 Immunologic Probe or Cytochemical Markers at Each Stage of Megakaryocyte and Platelet Maturation
BFU-Meg CFU-Meg LD-CFU-Meg MK-I MK-II MK-III Platelets
CD34; stem cell marker
HLA-DR
Peroxidase by cytochemical stain in transmission electron microscopy (CD41) glycoprotein IIb/IIIa (fibrinogen receptor)
by flow cytometry
CD42; glycoprotein Ib, part of VWF receptor, by flow cytometry
mpl (TPO receptor site) by flow cytometry
PF4 by flow cytometry
VWF by immunostaining
CD36; glycoprotein IV
Fibrinogen

BFU-Meg, Burst-forming unitmegakaryocyte; CFU-Meg, colony-forming unitmegakaryocyte; LD-CFU-Meg, low-density colony-forming unitmegakaryocyte; MK-I, megakaryo-
blast; MK-II, promegakaryocyte; MK-III, megakaryocyte; PF4, platelet factor 4; TPO, thrombopoietin; VWF, von Willebrand factor.
156 PART II Hematopoiesis

condensed with light and dark patches. The cytoplasm is azu- The proplatelet processes pierce through or between sinusoid-
rophilic (lavender), granular, and platelet-like, because of the lining endothelial cells, extend into the venous blood, and
spread of the DMS and -granules. At full maturation, platelet release platelets (Figure 13-8). Sometimes whole megakaryo-
shedding, or thrombocytopoiesis, proceeds. cytes escape the marrow in this fashion to lodge in other
organs, such as the lungs. Morphologists presume that throm-
Thrombocytopoiesis (Platelet Shedding) bopoiesis leaves behind naked megakaryocyte nuclei to be
Figure 13-7 appears to show platelet shedding. One cannot consumed by marrow macrophages, although these are rarely
find reliable evidence for platelet budding or shedding seen in bone marrow aspirate films. Their absence leads a few
simply by examining megakaryocytes in situ, even in well- morphologists to speculate that the lung is the primary site of
structured bone marrow biopsy preparations. In megakaryo- thrombopoiesis.13
cyte cultures examined by transmission electron microscopy,
the DMS dilates, longitudinal bundles of tubules form, cyto- Hormones and Cytokines of
plasmic extensions called proplatelet processes develop, and Megakaryocytopoiesis
transverse constrictions appear throughout the processes.12 TPO is a 70-kD molecule with 23% homology with erythro-
poietin.14 Messenger ribonucleic acid (RNA) for TPO has been
found in the kidney, liver, stromal cells, and smooth muscle
cells, though liver has the most copies. TPO circulates in plasma
and is the ligand that binds a megakaryocyte and platelet
membrane receptor protein, mpl, named for v-mpl, a viral
oncogene associated with murine myeloproliferative leukemia.
The concentration of TPO is inversely proportional to platelet
and megakaryocyte mass, which implies that membrane
binding and consequent disposal of TPO by platelets is the
primary control mechanism.15 Investigators have used in
vitro and in vivo experiments to show that TPO induces
stem cells to differentiate into megakaryocyte progenitors in
synergy with cytokines and that it further induces differentia-
tion of megakaryocyte progenitors into megakaryocytes,
induces the proliferation and maturation of megakaryocytes,
Figure 13-7 This image appears to illustrate a terminal megakaryocyte and induces platelet release (Table 13-3).16,17 Recombinant
shedding platelets; however, the appearance is more likely an artifact of film TPO in several forms elevates the platelet count in healthy
preparation. donors and in patients treated for a variety of neoplasms,

Megakaryocyte

Endothelial cells

Protoplatelet
process

Figure 13-8 Megakaryocyte is adjacent to the abluminal endothelial cell membrane and extends a proplatelet process through or between the endothelial
cells into the vascular sinus. (Modified from Powers LW: Diagnostic hematology: clinical and technical principles, St Louis, 1989, Mosby.)
CHAPTER 13 Platelet Production, Structure, and Function 157

TABLE 13-3 Hormones and Cytokines That Control Megakaryocytopoiesis


Differentiation to Differentiation to
Cytokine/Hormone Progenitors Megakaryocytes Late Maturation Thrombopoiesis Clinical Use
TPO + + + 0 Available
IL-3 + + 0
IL-6 0 0 + +
IL-11 0 + + + Available

Cytokines and hormones that have been shown to interact synergistically with TPO and IL-3 are IL-6 and IL-11; stem cell factor, also called kit ligand or mast cell growth factor;
granulocyte-macrophage colony-stimulating factor; granulocyte colony-stimulating factor; and erythropoietin.
Megakaryocyte inhibition: platelet factor 4, -thromboglobulin, neutrophil-activating peptide 2, IL-8.
IL, Interleukin; TPO, thrombopoietin.

including acute leukemia, and the commercial form NPlate makes them hard to examine for internal structure. In the
(romiplostim, Amgen) is effective in raising the platelet count blood, their surface is even, and they flow smoothly through
in immune thrombocytopenic purpura.18 veins, arteries, and capillaries. In contrast to leukocytes, which
Cell-derived stimulators of megakaryocytopoiesis include tend to roll along the vascular endothelium, platelets cluster
interleukin-3 (IL-3), IL-6, and IL-11. IL-3 seems to act in with the erythrocytes near the center of the blood vessel. Unlike
synergy with TPO to induce early differentiation of stem cells, erythrocytes, however, platelets move laterally with the leuko-
whereas IL-6 and IL-11 act in the presence of TPO to enhance cytes into the white pulp of the spleen, where both become
the later phenomena of endomitosis, megakaryocyte matura- sequestered.
tion, and platelet release. IL-11 has been synthesized and mar- The normal peripheral blood platelet count is 150 to 400
keted by Genetics Institute as Neumega (oprelvekin) and used 109/L. This count represents only two thirds of available plate-
to stimulate platelet production in patients with chemotherapy- lets, because the spleen sequesters an additional one third.
induced thrombocytopenia.19 Other cytokines and hormones Sequestered platelets are immediately available in times of
that participate synergistically with TPO and the interleukins demand, for example, in acute inflammation or after an injury,
are stem cell factor, also called kit ligand or mast cell growth factor; major surgery, or plateletpheresis. In hypersplenism or spleno-
granulocyte-macrophage colony-stimulating factor (GM-CSF); megaly, increased sequestration may cause a relative thrombo-
granulocyte colony-stimulating factor (G-CSF); and erythro- cytopenia. Under conditions of hemostatic need, platelets
poietin (EPO). The list continues to grow. answer cellular and humoral stimuli by becoming irregular and
Platelet factor 4 (PF4), -thromboglobulin, neutrophil- sticky, extending pseudopods, and adhering to neighboring
activating peptide 2, IL-8, and other factors inhibit in vitro structures or aggregating with each other.
megakaryocyte growth, which indicates that they may have a Reticulated platelets, sometimes known as stress platelets,
role in the control of megakaryocytopoiesis in vivo. Internally, appear in compensation for thrombocytopenia.21 Reticulated
reduction in the transcription factors FOG, GATA-1, and NF-E2 platelets are markedly larger than ordinary mature circulating
diminish megakaryocytopoiesis at the progenitor, endomito- platelets; their diameter in blood films exceeds 6m and their
sis, and terminal maturation phases.20 MPV reaches 12 to 14fL.22 Like ordinary platelets, they round
up in EDTA, but in citrated whole blood, reticulated platelets
are cylindrical and beaded, resembling megakaryocyte pro-
PLATELETS
platelet processes. Reticulated platelets carry free ribosomes
The proplatelet process sheds platelets, cells consisting of and fragments of rough endoplasmic reticulum, analogous to
granular cytoplasm with a membrane but no nuclear material, red blood cell reticulocytes, which has triggered speculation
into the venous sinus. Their diameter in the monolayer of a that they arise from early and rapid proplatelet extension and
Wright-stained peripheral blood wedge film averages 2.5m. release. Nucleic acid dyes such as thiazole orange bind the RNA
MPV, as measured in an isotonic suspension flowing through of the endoplasmic reticulum. This property is exploited by
the detector cell of a clinical profiling instrument, ranges profiling instruments to provide a quantitative evaluation of
from 8 to 10fL. A frequency distribution of platelet volume reticulated platelet production under stress, a measurement
is log-normal, however, which indicates a subpopulation of that may be more useful than the MPV.23 This measurement is
large platelets (see Figure 39-14). Volume heterogeneity in normal confounded by platelet dense granules, which falsely raise the
healthy humans reflects variation in platelet release volume and reticulated platelet count by taking up nucleic acid dyes.
is not a function of platelet age or vitality, as many authors have
previously assumed.5
PLATELET ULTRASTRUCTURE
Circulating, resting platelets are biconvex, although collec-
tion of blood using the anticoagulant ethylenediaminetetraace- Platelets, although anucleate, are strikingly complex and are
tic acid (EDTA) induces them to round up. On a Wright-stained metabolically active. Their ultrastructure has been studied
wedge-preparation blood film, platelets appear circular to using scanning and transmission electron microscopy, flow
irregular, lavender, and granular, although their small size cytometry, and molecular sequencing.
158 PART II Hematopoiesis

Carbohydrates Fatty acid


in plasma tails Esterified
cholesterol

Transmembranous glycoprotein Polar head


Microfilaments interacts with microfilaments groups
in cytoplasm

Figure 13-9 The platelet possesses a standard biologic membrane composed of a phospholipid bilayer, esterified cholesterol, and a series of transmem-
branous proteins.

Resting Platelet Plasma Membrane other plasma proteins, in many instances transporting them to
The platelet plasma membrane resembles any biologic storage organelles within in a process called endocytosis.
membrane: a bilayer composed of proteins and lipids, as At 20 to 30nm, the platelet glycocalyx is thicker than the
diagrammed in Figure 13-9. The predominant lipids are phos- analogous surface layer of leukocytes or erythrocytes. This thick
pholipids, which form the basic structure, and cholesterol, layer is adhesive and responds readily to hemostatic require-
which distributes asymmetrically throughout the phospholip- ments. The platelet literally carries its functional environment
ids. The phospholipids form a bilayer with their polar heads with it, meanwhile maintaining a negative surface charge that
oriented toward aqueous environmentstoward the plasma repels other platelets, other blood cells, and the endothelial
externally and the cytoplasm internally. Their fatty acid chains, cells that line the blood vessels.
esterified to carbons 1 and 2 of the phospholipid triglyceride The plasma membrane is selectively permeable, and
backbone, orient toward each other, perpendicular to the plane the membrane bilayer provides phospholipids that support
of the membrane, to form a hydrophobic barrier sandwiched platelet activation internally and plasma coagulation exter-
within the hydrophilic layers. nally. The anchored glycoproteins support essential plasma
The neutral phospholipids phosphatidylcholine and sphin- surfaceoriented glycosylated receptors that respond to cellular
gomyelin predominate in the plasma layer; the anionic or and humoral stimuli, called ligands or agonists, transmitting
polar phospholipids phosphatidylinositol, phosphatidyletha- their stimulus through the membrane to internal activation
nolamine, and phosphatidylserine predominate in the inner, organelles.
cytoplasmic layer. These phospholipids, especially phosphati-
dylinositol, support platelet activation by supplying arachi- Surface-Connected Canalicular System
donic acid, an unsaturated fatty acid that is converted to the The plasma membrane invades the platelet interior, producing
eicosanoids prostaglandin and thromboxane during platelet its unique surface-connected canalicular system (SCCS). The
activation. Phosphatidylserine flips to the outer surface on acti- SCCS twists spongelike throughout the platelet, enabling the
vation and is the phospholipid surface on which coagulation platelet to store additional quantities of the hemostatic
enzymes assemble.24 proteins of the glycocalyx, raising its capacity manyfold. The
Esterified cholesterol moves freely throughout the hydro- glycocalyx is less developed in the SCCS and lacks some of
phobic internal layer, exchanging with unesterified plasma the glycoprotein receptors present on the surface. The SCCS
cholesterol. Cholesterol stabilizes the membrane, maintains is the route for endocytosis and for secretion of granular
fluidity, and helps control the transmembranous passage of contents upon platelet activation.
materials.
Anchored within the membrane are glycoproteins and Dense Tubular System
proteoglycans; these support surface glycosaminoglycans, oli- Parallel and closely aligned to the SCCS is the dense tubular
gosaccharides, and glycolipids. The platelet membrane surface, system (DTS), a condensed remnant of the rough endoplasmic
called the glycocalyx, also absorbs albumin, fibrinogen, and reticulum. Having abandoned its usual protein production
CHAPTER 13 Platelet Production, Structure, and Function 159

function upon platelet release, the DTS sequesters Ca2+ adhesion. Another collagen-binding receptor is GP VI, a
and bears a series of enzymes that support platelet activation. member of the immunoglobulin gene family, so named because
These enzymes include phospholipase A2, cyclooxygenase, and the genes of its members have multiple immunoglobulin-like
thromboxane synthetase, which support prostaglandin synthe- domains. The unclassified platelet receptor GP IV also binds
sis, and phospholipase C, which supports production of ino- collagen and the adhesive protein thrombospondin.26
sitol triphosphate (IP3) and diacylglycerol (DAG). The DTS is Perhaps the most important adhesion receptor is GP Ib/
the control center for platelet activation. IX/V, a leucine-rich-repeat family CAM, named for its members
multiple leucine-rich domains. GP Ib/IX/V arises from the
Platelet Plasma Membrane Receptors That genes GP1BA, GP1BB, GP9, and GP5. It is composed of two
Provide for Adhesion molecules each of GP Ib, GP Ib, and GP IX, and one mol-
The platelet membrane supports more than 50 categories of ecule of GP V. This totals seven noncovalently-bound subunits.
receptors, including members of the cell adhesion molecule The two copies of subunit GP Ib account for VWF binding,
(CAM) integrin family, the CAM leucine-rich repeat family, the necessary in regions with blood flow shear rates higher
CAM immunoglobulin gene family, the CAM selectin family, the than 1000s1, as are found in capillaries and arterioles. Addi-
seven-transmembrane receptor (STR) family, and some miscella- tional sites on the GP Ib molecule bind thrombin. The
neous receptors.25 Table 13-4 lists the receptors that support accompanying GP Ib molecules cross the platelet membrane
the initial phases of platelet adhesion and aggregation. and interact with actin-binding protein to provide outside-in
Several integrins bind collagen, enabling the platelet to signaling. Two molecules of GP IX and one of GP V hold the
adhere to the injured blood vessel lining. Integrins are het- four GP Ib molecules together. Mutations in GP Ib, GP Ib,
erodimeric (composed of two dissimilar proteins) CAMs that or GP IX (but not GP V) are associated with a moderate-to-
integrate their ligands, which they bind on the outside of the severe mucocutaneous bleeding disorder, Bernard-Soulier syn-
cell, with the internal cytoskeleton, triggering activation. GP Ia/ drome. VWF deficiency is the basis for the most common
IIa, or 21, is an integrin that binds subendothelial collagen inherited bleeding disorder, von Willebrand disease. Von
in the damaged blood vessel wall to promote adhesion of the Willebrand disease also is associated with mucocutaneous
platelet to the vessel wall in regions of low blood flow, veins bleeding, although it is technically a plasma protein deficiency,
whose laminar flow shear rates are less than 1000s1. Likewise, not a platelet abnormality.27
51 and 61 bind the adhesive endothelial cell proteins The two subunits of GP IIb/IIIa, IIb and 3, are initially
laminin and fibronectin, which further promotes platelet inactive as they are distributed across the plasma membrane,

TABLE 13-4 Glycoprotein Platelet Membrane Receptors That Participate in Adhesion and the Initiation of Aggregation by
Binding Specific Ligands
Electrophoresis Current Cluster
Nomenclature Nomenclature Ligand Designation Comments
GP Ia/IIa Integrin: 21 Collagen CD29, CD49b
GP Ic/IIa Integrin: 51 Laminin CD29, CD49e
GP Ic/IIa Integrin: 61 Fibronectin CD29, CD49f
GP IV Miscellaneous platelet Collagen II and CD36 GP IV provides signal transduction to trigger aggregation.
receptor thrombospondin It is distributed on the plasma membrane, SCCS, and
20% on -granule membranes.
GP VI CAM of the Collagen
immunoglobulin
gene family
GP Ib/IX/V CAM of the leucine-rich VWF and thrombin CD42a, CD42b, GP Ib/IX/V is a 2:2:2:1 complex of GP Ib and Ib, GP
repeat family bind GP Ib; CD42c, CD42d IX, and GP V. There are 25,000 copies on the resting
thrombin cleaves platelet membrane surface, 5-10% on the -granule
a site on GP V membrane, but few on the SCCS membrane. GP Ib
is the VWF-specific site. Fifty percent of GP Ib/Ib is
cleared from the membrane on activation. Bernard-
Soulier syndrome mutations are identified for all but
GP V. Bound to subsurface actin-binding protein.
GP IIb/IIIa Integrin: IIb1 Fibrinogen, VWF CD41, CD61 GP IIb and IIIa are distributed on the surface membrane,
SCCS, and -granule membranes (30%). Heterodimer
forms on activation.

CAM, Cell adhesion molecule; GP, glycoprotein; SCCS, surface-connected canalicular system; VWF, von Willebrand factor.
160 PART II Hematopoiesis

the SCCS, and the internal layer of -granule membranes. These TABLE 13-5 STRs* for Thrombin, Adenosine
form an active heterodimer, IIb3, only when they encounter Diphosphate, Epinephrine, and the Prostaglandins
an inside-out signaling mechanism triggered by collagen
STR Receptor-Ligand Interaction
binding to GP VI or VWF binding to GP Ib/IX/V. Although
Coupled to Signaling
various agonists may activate the platelet, IIb3 is a physiologic
requisite because it binds fibrinogen, generating interplatelet Receptor Ligand G proteins
aggregation. Mutations in IIb or 3 cause a severe inherited PAR1 Thrombin Coupled to G1 protein that reduces
mucocutaneous bleeding disorder, Glanzmann thrombas cAMP; coupled to Gq and G12 proteins
thenia. The IIb3 integrin also binds VWF, vitronectin, and that increase IP3 and DAG
fibronectin, all adhesive proteins that share the target arginine- PAR4 Thrombin Coupled to Gq and G12 proteins that
glycine-aspartate (RGD) amino acid sequence with fibrinogen.28 increase IP3 and DAG
GP IIb/IIIa (IIb3) is the target receptor for the intravenous P2Y1 ADP Coupled to Gq protein that increases IP3
glycoprotein inhibitor (GPI) drugs ReoPro (abciximab, Eli and DAG
Lilly), Integrilin (eptifibatide, Millennium Pharmaceuticals) P2Y12 ADP Coupled to G1 protein that reduces cAMP
and Aggrastat (tirofiban, Medicure Pharma), any one of which TP and TP TXA2 Coupled to Gq protein that increases IP3
may be employed during cardiac catherization. Use of the and DAG
glycoprotein inhibitors requires platelet function assays for 2-adrenergic Epinephrine Coupled to G1 protein that reduces
efficacy and safety (see Chapter 46). cAMP; potentiates effects of ADP,
thrombin, and TXA2
Platelet Plasma Membrane Receptors IP PGI2 Coupled to GS protein that increases
That Provide for Activation: cAMP to inhibit activation
The Seven-Transmembrane Receptors
Thrombin, thrombin receptor activation peptide (TRAP), ADP, Adenosine diphosphate; cAMP, cyclic adenosine triphosphate; DAG, diacylglyc-
erol; IP3, inositol triphosphate; PAR, protease-activated receptor; PGI2, prostaglandin
adenosine diphosphate (ADP), epinephrine, and the prosta- I2 (prostacyclin) receptor; STR, seven-transmembrane receptor; TXA2, thromboxane A2;
glandin (eicosanoid, cyclooxygenase) pathway product throm- P2Y1 and P2Y12, members of the purigenic receptor family; TP and TP, thromboxane
boxane A2 (TXA2) all activate platelets. These platelet agonists receptors; IP, inositol receptor.
*Named for their peculiar sevenfold membrane anchorage, these receptors mediate
are ligands for STRs, so named for their unique membrane- outside-in platelet activation by signaling G proteins.
anchoring structure. The STRs have seven hydrophobic anchor-
ing domains supporting an external binding site and an
internal terminus that interacts with G proteins for outside-in cells. This interaction raises the internal cyclic adenosine
platelet signaling. The STRs are listed in Table 13-5.29 monophosphate (cAMP) concentration of the platelet and
Thrombin cleaves two STRs, protease-activated receptor 1 blocks activation. The platelet membrane also presents STRs
(PAR1) and PAR4, that together have a total of 1800 mem- for serotonin, platelet-activating factor, prostaglandin E2, PF4,
brane copies on an average platelet. Thrombin cleavage of and -thromboglobulin.
either of these two receptors activates the platelet through at
least two internal physiologic pathways, described later in the Additional Platelet Membrane Receptors
Platelet Activation section. Thrombin also interacts with plate- About 15 clinically relevant receptors were discussed in the
lets by binding or digesting two CAMs in the leucine-rich preceding paragraphs. The platelet supports many additional
repeat family, GP Ib (which is part of the GP Ib/IX/V VWF receptors. The CAM immunoglobulin family includes the
adhesion site) and GP V. ICAMs (CD50, CD54, CD102), which play a role in inflamma-
There are about 600 copies of the high-affinity ADP recep- tion and the immune reaction; PECAM (CD31), which medi-
tors P2Y1 and P2Y12. These STRs activate the platelet through ates platelettowhite blood cell and platelettoendothelial
the G-protein signaling pathways. Platelets are partially inacti- cell adhesion; and FcRIIA (CD32), a low-affinity receptor for
vated by prasugrel (Effient), clopidogrel (Plavix) and ticlopi- the immunoglobulin Fc portion that plays a role in a danger-
dine (Ticlid), drugs that occupy P2Y12 (see Chapter 46). These ous condition called heparin-induced thrombocytopenia (see
drugs are standard oral antithrombotic treatments prescribed Chapter 42).31 P-selectin (CD62) is an integrin that encourages
after acute myocardial infarctions, ischemic cerebrovascular platelets to bind endothelial cells, leukocytes, and each other.32
accidents, and venous thromboembolic events. Platelet func- P-selectin is found on the -granule membranes of the resting
tion assays are regularly employed to monitor prasugrel and platelet, but migrates via the SCCS to the surface of activated
clopidogrel efficacy and establish dosages.30 platelets. P-selectin or CD62 quantification by flow cytometry
TP and TP bind TXA2. This interaction produces more is a successful clinical means for measuring in vivo platelet
TXA2 from the platelet, an autocrine (self-perpetuating) system activation.
that activates neighboring platelets. Epinephrine binds 2-
adrenergic sites that couple to G proteins and open up mem- Platelet Cytoskeleton: Microfilaments
brane calcium channels. The 2-adrenergic sites function and Microtubules
similarly to those on heart muscle. Finally, the receptor site IP A thick circumferential bundle of microtubules maintains the
binds prostacyclin, a prostaglandin produced from endothelial platelets shape. The circumferential microtubules parallel the
CHAPTER 13 Platelet Production, Structure, and Function 161
plane of the disc and reside just within, although not touching, (Figure 13-10). The -granules are filled with proteins, some
the plasma membrane. There are 8 to 20 tubules composed of endocytosed, some synthesized within the megakaryocyte and
multiple subunits of tubulin that disassemble at refrigerator stored in platelets (Table 13-6). Several -granule proteins are
temperature or when treated with colchicine. When microtu- membrane bound. As the platelet becomes activated, -granule
bules disassemble, platelets become round, but upon warming membranes fuse with the SCCS. Their contents flow to the
to 37 C, they recover their original disc shape. On cross nearby microenvironment, where they participate in adhesion
section, microtubules are cylindrical, with a diameter of 25nm, and aggregation and support plasma coagulation.
and hollow. The circumferential microtubules could be a Gray platelet syndrome is an inherited absence of -granule
single spiral tubule.33 Besides maintaining platelet shape, contents. The granule membranes are present with their
microtubules contract on activation to encourage expression membrane-bound proteins, but the anticipated soluble pro-
of -granule contents. They also reassemble in long parallel teins normally within are missing. In gray platelet syndrome,
bundles to provide rigidity to pseudopods. platelets appear large and light gray in Wright-stained blood
In the narrow area between the microtubules and mem- films. Platelet aggregation in response to ADP, collagen, epi-
brane lies a thick meshwork of microfilaments composed nephrine, and thrombin is diminished, and patients experi-
of actin. Actin is contractile in platelets (as in muscle) and ence moderate to severe lifelong mucocutaneous bleeding.
anchors the plasma membrane glycoproteins and proteogly- There are two to seven -granules per platelet. Also called
cans. Actin also is present throughout the platelet cytoplasm, dense bodies, these granules appear later than -granules in
constituting 20% to 30% of platelet protein. In the resting megakaryocyte differentiation and stain black (opaque) when
platelet, actin is globular and amorphous, but as the cytoplas- treated with osmium in transmission electron microscopy.
mic calcium concentration rises, actin becomes filamentous Small molecules are probably endocytosed and are stored in
and contractile. the -granules; these are listed in Table 13-7. In contrast to the
The cytoplasm also contains intermediate filaments, rope- -granules, which employ the SCCS, -granules migrate to
like polymers 8 to 12nm in diameter, of desmin and vimentin. the plasma membrane and release their contents directly into
The intermediate filaments connect with actin and the tubules, the plasma upon platelet activation. Membranes of -granules
maintaining platelet shape. Microtubules, actin microfila- support the same integral proteins as the -granules
ments, and intermediate microfilaments control platelet shape P-selectin, GP IIb/IIIa, and GP Ib/IX/V, for instancewhich
change, extension of pseudopods, and secretion of granule implies a common source for the membranes of both types of
contents. granules.
Storage pool disorder is the name given to diminished -
Platelet Granules: -Granules, -Granules, granule contents. Storage pool disorders occur in inherited
and Lysosomes diseases characterized by albinism, such as Hermansky-Pudlak
There are 50 to 80 -granules in each platelet. In contrast to and Chdiak-Higashi syndromes, as well as in Wiskott-Aldrich
the nearly opaque -granules, -granules stain medium gray in syndrome, a nonalbinism X-linked recessive -granule defi-
osmium-dye transmission electron microscopy preparations ciency characterized by severe eczema. The inherited storage

Plasma
membrane
and glycocalyx

SCCS

granule

Microfilaments
SCCS

DTS
granule

Circumferential
microtubules
Figure 13-10 Ultrastructure of the circulating (resting) platelet. The biconvex shape is maintained by the circumferential microtubules. DTS, Dense tubular
system; SCCS, surface-connected canalicular system.
162 PART II Hematopoiesis

TABLE 13-6 Representative -Granule Proteins and -granules are reduced and abnormal in size, shape, and
contents. There may be mucocutaneous bleeding or even
Coagulation Noncoagulation
thrombosis.
Proteins Proteins
Storage pool disorder (-granule deficiency) does not
Proteins Present in Plasma and -Granules affect Wright stain platelet morphology, but platelets fail
Endocytosed Fibronectin Albumin to secrete when treated with thrombin or TRAP in lumiag-
Fibrinogen Immunoglobulins gregometry. Mepacrine, a quinidine-derived fluorescent dye,
Megakaryocyte synthesized Factor V stains -granule phosphates, which allows quantitation in flow
Thrombospondin cytometry.
VWF Refer to Chapter 44 for more information on platelet
granules, gray platelet syndrome, and storage pool disorder.
Present in -Granules, But Not Plasma Platelets also have a few lysosomes, similar to those in
(Megakaryocyte Synthesized) neutrophils, 300-nm diameter granules that stain positive for
-Thromboglobulin EGF arylsulfatase, -glucuronidase, acid phosphatase, and catalase.
HMWK Multimerin The lysosomes probably digest vessel wall matrix components
PAI-1 PDC1 during in vivo aggregation and may also digest autophagic
Plasminogen PDGF debris.
PF4 TGF-
Protein C inhibitor VEGF/VPF
PLATELET ACTIVATION
Membrane-Bound Proteins
Restricted to -granule P-selectin GMP33 Although the following discussion seems to imply a linear
membrane process, adhesion, aggregation, and secretion are often
Osteonectin simultaneous.34,35
In -granule and plasma GP IIb/IIIa cap1
membrane Adhesion: Platelets Reversibly Bind Elements
GP IV CD9 of the Vascular Matrix
GP Ib/IX/V PECAM-1 Platelets repair minor injuries to blood vessel linings, such
as regular sloughing of senescent endothelial cells, through
EGF, Endothelial growth factor; GMP, guanidine monophosphate; GP, glycoprotein; adhesion (Figures 13-11 and 13-12).36 Platelets may adhere
HMWK, high-molecular-weight kininogen; Ig, immunoglobulin; PAI-1, plasminogen
directly to collagen of the vascular matrix in veins or venules
activator inhibitor-1; PDCI, platelet-derived collagenase inhibitor; PDGF, platelet-
derived growth factor; PECAM-1, plateletendothelial cell adhesion molecule-1; PF4, through the CAMs described previously: GP Ia/IIa, GP IV,
platelet factor 4; TGF-, transforming growth factor-; VEGF/VPF, vascular endothelial and GP VI.
growth factor/vascular permeability factor; VWF, von Willebrand factor; cap1, adenyl
In capillaries and arterioles, where the blood flows rapidly
cyclaseassociated protein.
and the shear rate exceeds 1000s1, the site of injury is first
carpeted by VWF. VWF circulates as a multimeric globulin
TABLE 13-7 -Granule (Dense Body) Contents
with a molecular weight of between 800,000 D and 20 million
Small Molecule Comment D, one of the largest proteins in the plasma. The combination
of injury and shear stress unrolls the globular molecule to
ADP Nonmetabolic, supports neighboring platelet
form fibrillar strands that coat injury sites and bind subendo-
aggregation by binding to ADP receptors
thelial collagen. A liver-secreted plasma enzyme, VWF-cleaving
P2Y1, P2Y12
protease, controls thrombogenicity by rapidly digesting fibrillar
ATP Function unknown, but ATP release is detected
VWF to inactive fragments.37 Platelets adhere to fibrillar VWF
using firefly luciferase luminescence as an
through their GP Ib/IX/V receptor, and the VWF-platelet layer
in vitro measure of platelet activationa
fills in the space made by the injury to await replacement
method called lumiaggregometry
by normal vascular cells (Figure 13-13). Although seldom an
Serotonin Vasoconstrictor that binds endothelial cells and
isolated process, adhesion alone may exclude secretion of
platelet membranes
granule contents or the energy-dependent redistribution of
Ca2+ and Mg2+ Divalent cations support platelet activation and
CAMs such as GP IIb/IIIa (IIb3) or P-selectin. Adhesion does
coagulation
require contraction of actin molecules, however, and the
ADP, Adenosine diphosphate; ATP, adenosine triphosphate; P2Y1 and P2Y12, members formation of pseudopods.
of the purigenic receptor family. Although history records several attempts, no one has
devised a clinical laboratory method that isolates and measures
pool disorders are associated with mild mucocutaneous platelet adhesion without simultaneously involving aggrega-
bleeding. tion and secretion. Clinicians routinely employ laboratory
More commonly, storage pool disorder arises with irregular instrumentation to measure VWF activity and concentration,
platelet production in myeloproliferative neoplasms or myelodys- however, and clinicians may quantitate several CAMs through
plastic syndromes. In these hematologic diseases, both -granules flow cytometry in specialized laboratories.
CHAPTER 13 Platelet Production, Structure, and Function 163

Collagen

Tunica
media
SMC FB SMC SMC

IEL
Tunica
EC EC EC EC EC EC EC EC
intima

RBC PLT
RBC PLT RBC
PLT
WBC

EC EC EC EC EC EC EC EC Normal intimae suppress


hemostasis:
Prostacyclin
Heparan sulfate
SMC FB SMC FB SMC Tissue factor pathway inhibitor
Nitric oxide
Thrombomodulin
Figure 13-11 Normal blood flow in intact vessels. Red blood cells (RBCs) and platelets (PLTs) flow near the center; white blood cells (WBCs) marginate.
EC, Endothelial cell; FB, fibroblast; SMC, smooth muscle cell.

Collagen

Tunica
media
SMC FB SMC SMC

VWF
IEL PLT
Tunica
EC EC PLT PLT EC EC EC EC
intima

RBC PLT
RBC PLT RBC
PLT
WBC

EC EC EC EC EC EC EC EC

SMC FB SMC FB SMC

Figure 13-12 In capillaries and arterioles, von Willebrand factor (VWF) supports platelet adhesion as it carpets an injury and binds collagen. Platelets
(PLTs) bind VWF to effect vessel lining repair. In veins, platelets bind collagen directly (not pictured). EC, Endothelial cell; FB, fibroblast; RBC, red blood cell;
SMC, smooth muscle cell.

Aggregation: Platelets Irreversibly Bind cytoplasm forms as the platelet exhausts internal energy
Each Other sources. Aggregation is a part of primary hemostasis, and its
When vessel damage is extensive, platelets adhere and aggre- irreversible end point is the white clot or platelet-VWF plug.
gate (Figures 13-14 and 13-15). Aggregation requires the active Although a normal part of vessel repair, white clots often imply
conformation of the GP IIb/IIIa (IIb3) integrin and includes inappropriate platelet activation in uninjured arterioles and
pseudopod formation and redistribution of P-selectin to the arteries and are the pathologic basis for arterial thrombotic
surface membrane (Figure 13-16). Membrane phospholipids events, such as acute myocardial infarction, peripheral vascular
also redeploy, with the more polar molecules, such as phos- disease, and strokes. Except for VWF, coagulation proteins are
phatidylserine, flipping to the outer layer. GP IIb/IIIa binds the excluded from white clots.
RGD sequence of any plasma protein; the protein most readily The combination of polar phospholipid exposure, platelet
available is fibrinogen, which is the major player in aggrega- microparticle dispersion, and secretion of the platelets -
tion. Membrane integrity is lost, and a syncytium of platelet and -granule contents triggers secondary hemostasis, called
164 PART II Hematopoiesis

Collagen
binding site: GP
la/lla (CD 49b),
GP lV and Vl

PLT
VWF Factor VlII
binding functional site
site
GP lb/IX/V
CD 42
VWF Vlll

Internal elastic lamina

SMC FB SMC FB SMC

VWF collagen
binding site Collagen

Figure 13-13 Von Willebrand factor (VWF) binds subendothelial collagen in an injured blood vessel. Platelets (PLTs) bind VWF through the binding site
glycoprotein (GP) Ib/IX/V (CD 42). Platelets also bind collagen directly. VWF binds and stabilizes factor VIII. EC, Endothelial cell; FB, fibroblast; SMC, smooth
muscle cell.

Tunica
mediae
SMC FB SMC SMC

Internal elastic lamina


Tunica
EC EC EC EC EC EC EC EC
intima

RBC PLT
RBC PLT RBC
PLT
WBC

EC EC EC EC EC EC EC EC Damaged intimae trigger


hemostasis:
Phospholipids
SMC FB SMC FB SMC Collagen
Tissue factor
VWF

Figure 13-14 Trauma to the blood vessel wall exposes collagen and tissue factor, triggering platelet adhesion and aggregation. EC, Endothelial cell;
FB, fibroblast; PLT, platelet; RBC, red blood cell; SMC, smooth muscle cell.

coagulation (see Chapter 40). Fibrin and red blood cells deposit Secretion: Activated Platelets Release
around and within the platelet syncytium, forming a bulky Granular Contents
red clot. The red clot is essential to wound repair, but is Outside-in platelet activation through ligand (agonist) binding
characteristic of inappropriate coagulation in venules and to integrins and STRs triggers actin microfilament contraction.
veins, resulting in deep vein thrombosis and pulmonary Intermediate filaments and microtubules contract, compress-
emboli. ing granules. Contents of -granules and lysosomes flow
Specialty and tertiary care coagulation laboratories provide through the SCCS; meanwhile -granule contents are secreted
in vitro whole blood or plasma platelet aggregometry and through the plasma membrane (Figure 13-17). The -granule
lumiaggregometry as a means for detecting and identifying contents are vasoconstrictors and platelet agonists that amplify
aggregation abnormalities. The bleeding time test, although primary hemostasis; several of the -granule contents are
still used, has limited predictive value. Several systems for the coagulation proteins (Table 13-8).
physicians office or near-patient laboratory, such as the Siemens By presenting polar phospholipids on their membrane sur-
PFA-100 (Deerfield, Ill.), and the Chronolog WBA (Havertown, faces, platelets provide a localized cellular milieu that supports
Pa.), also are available to test for platelet function.38 coagulation. Phosphatidylserine is the phospholipid on which
CHAPTER 13 Platelet Production, Structure, and Function 165

Platelets

EC

Internal elastic lamina

VWF
VWF
Collagen Collagen

Figure 13-15 Blood enters the wound and platelets adhere directly to collagen or to von Willebrand factor (VWF), triggering adhesion and aggregation.
EC, Endothelial cell. (Courtesy Kathy Jacobs, Chronolog, Inc., Havertown, Pa).
ADP

Resting
PLT

P2Y1
receptor
GP Ilb GP Illa
CD41 CD61

P-selectin
Activated
CD62 PLT GP Ilb/Illa
detection by
PAC-1

Figure 13-16 Activation of platelets (PLTs) by an agonist (ligand) such as adenosine diphosphate (ADP) generates an inside-out signal that supports
assembly of glycoprotein (GP) IIb/IIIa, detected by the polyclonal antibody PAC-1, and the appearance of P-selectin (CD62) on the membrane.

two coagulation pathway complexes form: factor IX/VIII hemostasis. Some proteins listed in Table 13-8 do not appear
(tenase) and factor X/V (prothrombinase), supported by ionic in Table 13-5. Neither list is exhaustive, because more -granule
calcium secreted by the -granules. The -granule contents contents are being identified.
fibrinogen, factors V and VIII, and VWF (which binds and There are several clinical laboratory assays of platelet secre-
stabilizes factor VIII) are secreted and increase the localized tions. The most successful is lumiaggregometry, which employs
concentrations of these essential coagulation proteins, which firefly luciferase to identify adenosine triphosphate secreted
further supports the action of tenase and prothrombinase. from -granules through luminescence. Immunoassays of PF4
Platelet secretions serve cell-based, controlled, localized coagu- or -thromboglobulin, unique to platelets and secreted by
lation. Table 13-8 lists some additional -granule secretions -granules, are available, but only from reference laboratories
that, although not proteins of the coagulation pathway, support using specialized blood specimen collection techniques. This
166 PART II Hematopoiesis

Forms White Clot or Platelet Plug


Contents of -granules Contents of -granules
Proteins: Fibrinogen, VWF, factors V,
Small molecules: Vlll, PF4, -TG, PDGF, P-selectin,
serotonin, ADP, ATP, Ca2+ many more

Thrombin

VWF
Tissue factor

Platelet plug (white dot) composed


of platelets and VWF
Figure 13-17 Platelets adhere, aggregate, and release -granule contents; serotonin is a vasoconstrictor and adenosine diphosphate (ADP) activates
neighboring platelets. Contents of the -granules, are released simultaneously. These are predominantly coagulation factors, but also the platelet-specific
factors platelet factor 4 (PF4), platelet-derived growth factor (PDGF), and -thromboglobulin (-TG). ATP, Adenosine triphosphate; VWF, von Willebrand factor.
(Courtesy Kathy Jacobs, Chronolog, Inc., Havertown, Pa).

is because uncontrolled ex vivo platelet activation factitiously


elevates the levels of these substances in specimens that are
collected and handled using routine processes.
TABLE 13-8 Selected -Granule Proteins and
Their Properties PLATELET ACTIVATION PATHWAYS
-Granule Protein Properties G Proteins
Platelet-derived growth factor Supports mitosis of vascular fibroblasts G proteins control cellular activation for all cells (not just
and smooth muscle cells platelets) at the inner membrane surface (Figure 13-18). G
Endothelial growth factor Supports mitosis of vascular fibroblasts proteins are heterotrimers that bind guanosine dip
and smooth muscle cells hosphate (GDP) when inactive. Membrane receptor-ligand
Transforming growth factor- Supports mitosis of vascular fibroblasts (agonist) binding promotes GDP release and its replacement
and smooth muscle cells with guanosine triphosphate (GTP). The G monomer briefly
Fibronectin Adhesion molecule disassociates, exerts guanosine triphosphatase activity, and
Thrombospondin Adhesion molecule hydrolyzes the bound GTP to GDP, releasing a phosphate
radical. The G protein resumes the resting state, but the hydro-
Platelet factor 4
-thromboglobulin } { Heparin neutralization
Found nowhere but platelet -granules lysis step provides the necessary phosphorylation trigger to
Plasminogen Fibrinolysis promotion and control energize the eicosanoid synthesis or the IP3-DAG pathway
Plasminogen activation Fibrinolysis promotion and control (Table 13-9).
inhibitor-1
2-Antiplasmin Fibrinolysis promotion and control Eicosanoid Synthesis
Protein C inhibitor Coagulation control The eicosanoid synthesis pathway, alternatively called the prosta-
glandin, cyclooxygenase, or thromboxane pathway, is one of two
CHAPTER 13 Platelet Production, Structure, and Function 167

1. Ligand Ligand

Receptor
Receptor

Inactive 2. Active
G-protein GDP G-protein 3. GTP

Zymogen 4. Enzyme

Figure 13-18 G-protein mechanism. 1. A ligand (agonist) binds its corresponding receptor. 2. G-protein swaps guanosine diphosphate (GDP) for guanosine
triphosphate (GTP). 3. GTP donates a high-energy phosphate radical to a zymogen and remains attached to the G-protein as GDP. 4. The zymogen is activated.
The G-protein returns to a resting state bound to GDP.

TABLE 13-9 G Proteins in Platelets and Their Functions


G Protein Coupled to Receptor Agonist (Ligand) Action Outcome
G1 PAR1 Thrombin Decelerate adenylate cyclase Reduce cAMP concentration
P2Y12 ADP
2-Adrenergic Epinephrine
Gq PAR1 Thrombin Activate phospholipase C Increase IP3-DAG
PAR4 Thrombin
P2Y1 ADP
TP and TP TXA2
G12 PAR1 Thrombin Activate protein kinase C Activate pleckstrin, actin microfilaments
PAR4 Thrombin
P2Y1 ADP
TP and TP TXA2
Gs IP Prostacyclin Accelerate adenylate cyclase Increase cAMP concentration

ADP, Adenosine diphosphate; cAMP, cyclic adenosine triphosphate; DAG, diacylglycerol; IP3, inositol triphosphate; PAR, protease-activated receptor; TXA2, thromboxane A2;
P2Y1 and P2Y12, members of the purigenic receptor family; TP and TP, thromboxane receptors; IP, inositol receptor.

essential platelet activation pathways triggered by G protein receptors TP or TP, decelerating adenylate cyclase activity
(Figure 13-19). The platelet membranes inner leaflet is rich and reducing cAMP concentrations, which mobilizes ionic
in phosphatidylinositol, a phospholipid whose number 2 calcium from the DTS (Figure 13-20). The rising cytoplasmic
carbon binds numerous unsaturated fatty acids, including calcium level causes contraction of actin microfilaments and
5,8,11,14-eicosatetraenoic acid, commonly called arachidonic platelet activation.
acid. Membrane receptor-ligand binding and the consequent The cyclooxygenase pathway in endothelial cells incorpo-
G-protein activation triggers phospholipase A2, a membrane rates the enzyme prostacyclin synthetase in place of the throm-
enzyme that cleaves the ester bond connecting the number 2 boxane synthetase in platelets. The eicosanoid pathway end
carbon of the triglyceride backbone with arachidonic acid. point for the endothelial cell is prostaglandin I2, or prostacy-
Cleavage releases arachidonic acid to the cytoplasm, where it clin, which binds the platelet IP receptor site. Prostacyclin
becomes the substrate for cyclooxygenase, anchored in the DTS. binding accelerates adenylate cyclase, increasing cAMP, and
Cyclooxygenase converts arachidonic acid to prostaglandin G2 moves ionic calcium to DTS sequestration. The endothelial cell
and prostaglandin H2, then thromboxane synthetase acts on pathway controls platelet activation in the intact blood vessel
prostaglandin H2 to produce TXA2. TXA2 binds membrane through this mechanism.
168 PART II Hematopoiesis

Phosphatidylinositol
ADP
Thrombin Phospholipase A2
Collagen R
Epinephrine
Arachidonic acid

PLT
Arachidonic

PLTdense
Cyclooxygenase
acid

densetubular
PgG2

tubularsystem
Peroxidase

PgH2

system
Thromboxane
synthase
TXA2 activates
platelet

Prostacyclin
synthase in
endothelial cell

Figure 13-19 Eicosanoid synthesis. The ligands (agonists) adenosine diphosphate (ADP), thrombin, collagen, or epinephrine bind their respective membrane
receptors (R). The combination activates phospholipase A2 through the G-protein mechanism described in Figure 13-18. Phospholipase A2 releases arachidonic
acid from membrane phosphatidylinositol. Arachidonic acid is acted upon by cyclooxygenase, peroxidase, and thromboxane synthase to produce thromboxane
A2 (TXA2), which activates the platelet through adenylate cyclase as shown in Figure 13-20. When reagent arachidonic acid is used as an agonist, it bypasses
the membrane and directly enters the eicosanoid pathway. In the endothelial cell the eicosanoid pathway is nearly identical, except that prostacyclin synthase
replaces thromboxane synthase. PgG2, Prostaglandin G2; PgH2, prostaglandin H2; PLT, platelet.

DTS
Ca2+

Ca2+ Ca2+
TXA2 suppresses
adenylate Ca2+ Active
Ca2+
cyclase Ca2+

ATP Decreased
cylic AMP

Ca2+ DTS Ca2+


Ca2+ Ca2+ Ca2+ Inactive
Ca2+
Figure 13-20 Second messenger system. In the platelet, thromboxane A2 (TXA2) suppresses adenylate cyclase and reduces cyclic adenosine monophos-
phate (AMP) concentration. This allows the release of ionic calcium from the dense tubular system (DTS). Ionic calcium supports contraction of actin microfila-
ments, which activates the platelet. In the endothelial cell prostacyclin (not pictured) has the opposite effect on adenylate cyclase, raising cyclic AMP and
sequestering ionic calcium in the DTS. ATP, Adenosine triphosphate.

Deficiencies in calcium mobilization, cyclooxygenase, and the response to low-concentration (1mg/mL) collagen,
thromboxane synthetase activity reduce TXA2 production and whereas aspirin-like disorders reduce aggregation response at
platelet activation, an inherited set of conditions identified both low and high (5mg/mL) concentrations of collagen.
collectively as platelet release disorder or aspirin-like disorder, TXA2 has a half-life of 30 seconds, diffuses from the platelet,
which is associated with mucocutaneous bleeding and intra- and spontaneously reduces to thromboxane B2, a stable, mea-
cranial hemorrhages. Aggregometry patterns in these disorders surable plasma metabolite. Efforts to produce a clinical assay
mimic the effect of aspirin, which causes a reduced response for plasma thromboxane B2 have been unsuccessful, because
to ADP, arachidonic acid, and epinephrine. Aspirin also reduces special specimen management is required to prevent ex vivo
CHAPTER 13 Platelet Production, Structure, and Function 169

Direct actin
contraction

Releases Ca2+
from DTS
C1HR1
DAG C2H-R2 O
C3HOPO
O_ IP3

PLC
O O
OPO OPO
O O

Figure 13-21 Phospholipase C (PLC) is activated by the G-protein mechanism and cleaves the phosphodiester bond from carbon 3 of phosphatidylinositol.
One product is diacylglycerol (DAG), which directly generates actin microfilament contraction. The other product is inositol triphosphate (IP3), which releases
ionic calcium from the dense tubular system (DTS). R1, Saturated fatty acid that remains attached to carbon 1; R2, unsaturated fatty acid that remains attached
to carbon 2. (Courtesy Larry D. Brace, PhD, Edward Hospital, Naperville, Ill.)

platelet activation with factitious unregulated release of throm- activation. IP3 promotes release of ionic calcium from the DTS,
boxane B2. Thromboxane B2 is acted on by a variety of liver which triggers actin microfilament contraction. IP3 also may
enzymes to produce an array of soluble urine metabolites, activate phospholipase A2. DAG triggers a multistep process:
including 11-dehydrothromboxane B2, which is stable and activation of phosphokinase C, which triggers phosphorylation
measurable.39 Immunoassays of urine 11-dehydrothromboxane of the protein pleckstrin, which regulates actin microfilament
B2 are used to characterize in vivo platelet activation and the contraction.
suppressing effect of aspirin.40 Internal platelet activation pathways, like internal pathways
of all metabolically active cells, are often called second messen-
gers because they are triggered by a primary ligand-receptor
Inositol TriphosphateDiacylglycerol binding event. Second messengers include G proteins, the eico-
Activation Pathway sanoid synthesis pathway, the IP3-DAG pathway, adenylate
The IP3-DAG pathway is the second G proteindependent cyclase, cAMP, and intracellular ionic calcium. This discussion
platelet activation pathway (Figure 13-21). G-protein activa- has been limited to activation pathways whose aberration
tion triggers the enzyme phospholipase C. Phospholipase C cause hemostatic disease. The reader is referred to cell physio
cleaves membrane phosphatidylinositol 4,5-bisphosphate to logy texts for a comprehensive discussion of cellular activation
form IP3 and DAG, both second messengers for intracellular pathways.

SUMMARY
Platelets arise from bone marrow megakaryocytes, which reside The platelet membrane supports many receptor sites that control
near the venous sinusoid. Megakaryocyte progenitors are recruited platelet activation on binding their respective ligands.
by IL-3, IL-6, IL-11, and TPO and mature via endomitosis. Platelets Platelets adhere to foreign surfaces, aggregate with each other,
are released into the bone marrow through shedding from pro- and secrete the substances stored within their granules.
platelet processes, a process called thrombopoiesis. Platelet activation is internally managed through G proteins, the
Circulating platelets are complex anucleate cells with a thick gly- eicosanoid synthesis pathway, and the IP3-DAG pathway.
cocalyx bearing an assortment of coagulation factors, extended by
the SCCS. Inside is a system of microfibrils and microtubules that Now that you have completed this chapter, go back and
accomplish platelet contraction and pseudopod extension and read again the case study at the beginning and respond
the granules that provide coagulation factors and vasoactive to the questions presented.
chemicals.
170 PART II Hematopoiesis

R E V I E W Q UESTIONS
1. The megakaryocyte progenitor that undergoes endomito- 6. What is the name of the eicosanoid metabolite
sis is: produced from endothelial cells that suppresses platelet
a. MK-I activity?
b. BFU-Meg a. TXA2
c. CFU-Meg b. Arachidonic acid
d. LD-CFU-Meg c. Cyclooxygenase
d. Prostacyclin
2. The growth factor that is produced in the kidney and
induces growth and differentiation of committed mega- 7. Which of the following molecules is stored in platelet
karyocyte progenitors is: -granules (dense bodies)?
a. IL-3 a. Serotonin
b. IL-6 b. Fibrinogen
c. IL-11 c. PF4
d. TPO d. Platelet-derived growth factor

3. What platelet organelle sequesters ionic calcium and binds 8. What plasma protein is essential to platelet adhesion?
a series of enzymes of the eicosanoid pathway? a. VWF
a. G protein b. Factor VIII
b. Dense granules c. Fibrinogen
c. DTS d. P-selectin
d. SCCS
9. Reticulated platelets can be enumerated in peripheral
4. What platelet membrane receptor binds fibrinogen and blood to detect:
supports platelet aggregation? a. Impaired production in disease states
a. GP Ib/IX/V b. Abnormal organelles associated with diseases such as
b. GP IIb/IIIa leukemia
c. GP Ia/IIa c. Increased platelet production in response to need
d. P2Y1 d. Inadequate rates of membrane cholesterol exchange
with the plasma
5. What platelet membrane phospholipid flips from the
inner surface to the plasma surface on activation and 10. Platelet adhesion refers to platelets:
serves as the assembly point for coagulation factors? a. Sticking to other platelets
a. Phosphatidylethanolamine b. Releasing platelet granule constituents
b. Phosphatidylinositol c. Providing the surface for assembly of coagulation
c. Phosphatidylcholine factors
d. Phosphatidylserine d. Sticking to surfaces such as subendothelial collagen

REFERENCES
1. Butkiewicz AM, Kemona H, Dymicka-Piekarska V, et al: Plate- 7. Cramer EM, Vainchenker W: Platelet production: cellular
let count, mean platelet volume and thrombocytopoietic and molecular regulation. In Colman RW, Marder VJ,
indices in healthy women and men. Thromb Res 118:199-204, Clowes AM, et al, editors: Hemostasis and thrombosis,
2006. Epub September 1, 2005. ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins,
2. Tsiara S, Elisaf M, Jagroop IA, et al: Platelets as predictors pp 443-462.
of vascular risk: is there a practical index of platelet activity? 8. Cramer EM, Fontenay M: Platelets: structure related to
Clin Appl Thromb Hemost 9:177-190, 2003. function. In Colman RW, Marder VJ, Clowes AM, et al,
3. Lichtman MA, Chamberlain JK, Simon W, et al: Parasinusoid editors: Hemostasis and thrombosis, ed 5, Philadelphia, 2006,
allocation of megakaryocytes in marrow. Am J Hematol 4:303- Lippincott Williams & Wilkins, pp 463-482.
312, 1978. 9. George JN, Colman RW: Overview of platelet structure and
4. Samii K, Pasteur E: Images in hematology. Emperipolesis, Am function. In Colman RW, Marder VJ, Clowes AM, et al,
J Hematol 59:64, 1998. editors: Hemostasis and thrombosis, ed 5, Philadelphia, 2006,
5. Kosaki G: In vivo platelet production from mature mega- Lippincott Williams & Wilkins, pp 437-442.
karyocytes: does platelet release occur via proplatelets? Int J 10. Kaushansky K: Megakaryopoiesis and thrombopoiesis. In
Hematol 81:208-219, 2005. Lichtman MA, Beutler E, Kills T, et al: Williams hematology,
6. Chang Y, Bluteau D, Bebili N, et al: From hematopoietic stem ed 7, New York, 2006, McGraw-Hill, pp 1571-1586.
cells to platelets. J Thrombos Haemostas 5(suppl 1):318-327, 11. Choi ES, Hokom M, Bartley T, et al: Recombinant human
2007. megakaryocyte growth and development factor (rHuMGDF),
CHAPTER 13 Platelet Production, Structure, and Function 171
a ligand for c-Mpl, produces functional human platelets in and thrombosis, ed 5, Philadelphia, 2006, Lippincott Williams
vitro. Stem Cells 13:317-322, 1995. & Wilkins, pp 547-554.
12. Patel SR, Richardson JL, Schulze H, et al: Differential roles of 26. Tandon NN, Kraslisz U, Jamieson GA: Identification of
microtubule assembly and sliding in proplatelet formation glycoprotein IV (CD36) as a primary receptor for platelet-
by megakaryocytes. Blood 106:4076-4085, 2005. collagen adhesion. J Biol Chem 264:7576-7583, 1989.
13. Levine RF, Eldor A, Shoff PK, et al: Circulating megakaryo- 27. Rivera J, Lozano ML, Corral J, et al: Platelet GP Ib/IX/V
cytes: delivery of large numbers of intact, mature megakaryo- complex: physiological role. J Physiol Biochem 56:355-365,
cytes to the lung. Eur J Haematol 51:233-246, 1993. 2000.
14. Kunicki TJ: Platelet glycoprotein polymorphisms and 28. Rao AK: Disorders of platelet function. In Kitchens CS,
relationship to function, immunogenicity, and disease. In Alving BM, Kessler CM, editors: Consultative hemostasis and
Colman RW, Marder VJ, Clowes AM, et al, editors: Hemostasis thrombosis, Philadelphia, 2002, Saunders, pp 133-148.
and thrombosis, ed 5, Philadelphia, 2006, Lippincott Williams 29. Jackson SP, Nesbitt WS, Kulkarni S: Signaling events underly-
& Wilkins, pp 493-506. ing thrombus formation. J Thromb Haemost 1:1602-1612,
15. Emmons RVB, Reid DM, Cohen R, et al: Human thrombo- 2003.
poietin levels are high when thrombocytopenia is due to 30. Moliterno DJ: Advances in antiplatelet therapy for ACS and
megakaryocyte deficiency and low when due to platelet PCI. J Interv Cardiol 21(suppl 1):S18-S24, 2008.
destruction. Blood 87:4068-4071, 1996. 31. Walenga JM, Jeske WP, Prechel MM, et al: Newer insights on
16. McDonald TP: Thrombopoietin: its biology, clinical aspects, the mechanism of heparin-induced thrombocytopenia.
and possibilities. Am J Pediatr Hematol Oncol 14:8-21, 1992. Semin Thromb Hemost 30(suppl 1):57-67, 2004.
17. Stasi R: Therapeutic strategies for hepatitis- and other 32. Keating FK, Dauerman HL, Whitaker DA, et al: Increased
infection-related immune thrombocytopenias. Semin Hematol expression of platelet P-selectin and formation of platelet-
46(1 suppl 2):S15-S25, 2009. leukocyte aggregates in blood from patients treated with
18. Rice L: Drug evaluation: AMG-531 for the treatment of throm- unfractionated heparin plus eptifibatide compared with
bocytopenias. Curr Opin Investig Drugs 7:834-841, 2006. bivalirudin. Thromb Res 118:361-369, 2006.
19. Sitaraman SV, Gewirtz AT: Oprelvekin. Genetics Institute. 33. White JG, Rao GH: Microtubule coils versus the surface mem-
Curr Opin Investig Drugs 2:1395-1400, 2001. brane cytoskeleton in maintenance and restoration of platelet
20. Chang M, Nakagawa PA, Williams SA, et al: Immune throm- discoid shape. Am J Pathol 152:597-609, 1998.
bocytopenic purpura (ITP) plasma and purified ITP mono- 34. Abrams CS, Brass LF: Platelet signal transduction. In Colman
clonal autoantibodies inhibit megakaryocytopoiesis in vitro. RW, Marder VJ, Clowes AM, et al, editors: Hemostasis and
Blood 102:887-895, 2003. thrombosis, ed 5, Philadelphia, 2006, Lippincott Williams &
21. Briggs C, Harrison P, Machin SJ: Continuing developments Wilkins, pp 617-631.
with the automated platelet count. Int J Lab Hematol 35. Abrams CS: Intracellular signaling in platelets. Curr Opin
29:77-91, 2007. Hematol 12:401-405, 2005.
22. Abe Y, Wada H, Sakakura M, et al: Usefulness of fully auto- 36. Tailor A, Cooper D, Granger DN: Platelet-vessel wall interac-
mated measurement of reticulated platelets using whole tions in the microcirculation. Microcirculation 12:275-285,
blood. Clin Appl Thromb Hemost 11:263-270, 2005. 2005.
23. Briggs C, Kunka S, Hart D, et al: Assessment of an immature 37. Levy GG, Motto DG, Ginsburg D: ADAMTS13 turns 3. Blood
platelet fraction (IPF) in peripheral thrombocytopenia. Br J 106:11-17, 2005.
Haematol 126:93-99, 2004. 38. Michelson AD: Methods for the measurement of platelet
24. Zieseniss S, Zahler S, Muller I, et al: Modified phosphatidyl- function. Am J Cardiol 103(suppl 3):20A-26A, 2009.
ethanolamine as the active component of oxidized low 39. Fritsma GA, Ens GE, Alvord MA, et al: Monitoring the anti-
density lipoprotein promoting platelet prothrombinase activ- platelet action of aspirin. JAAPA 14:57-62, 2001.
ity. J Biol Chem 276:19828-19835, 2001. 40. Eikelboom JW, Hankey GJ: Failure of aspirin to prevent ath-
25. Kunapuli SP: Platelet adenosine 5-diphosphate receptors. In erothrombosis: potential mechanisms and implications for
Colman RW, Marder VJ, Clowes AM, et al, editors: Hemostasis clinical practice. Am J Cardiovasc Drugs 4:57-67, 2004.
PART III Routine Laboratory Evaluation of Blood Cells

14 Routine and Point-of-Care Testing


in Hematology: Manual and
Semiautomated Methods
Karen S. Clark and Teresa G. Hippel

OUTLINE OBJECTIVES
Manual Cell Counts After completion of this chapter, the reader will be able to:
Equipment
Calculations 1. State the dimensions of the counting area of a 10. Classify erythrocytes according to size and hemoglo
White Blood Cell Count Neubauer ruled hemacytometer. bin content, using results of red blood cell (RBC)
Platelet Count 2. Describe the performance of manual cell counts for indices.
Erythrocyte Counts leukocytes, erythrocytes, and platelets, including 11. Describe the principle and procedure for performing
Body Fluid Cell Counts types of diluting fluids, typical dilutions, and typical a manual reticulocyte count and the clinical value of
Hemoglobin areas counted in the hemacytometer. the test.
Determination 3. Calculate dilutions for cell counts when given appro 12. Calculate the relative, absolute, and corrected reticu
Principle priate data. locyte values and a reticulocyte production index.
Microhematocrit 4. Calculate hemacytometer cell counts when given 13. Interpret the results of reticulocyte calculations to
Rule of Three
numbers of cells, area counted, and dilution. evaluate the degree of bone marrow erythropoiesis.
Red Blood Cell Indices
5. Describe the principle of cyanmethemoglobin assay 14. Describe the procedure for performing Westergren
Mean Cell Volume
Mean Cell Hemoglobin for determination of hemoglobin. erythrocyte sedimentation rates (ESRs).
Mean Cell Hemoglobin 6. Calculate the values for a standard curve for 15. State the diagnostic value of ESRs.
Concentration cyanmethemoglobin determination when given the 16. Correct white blood cell (WBC) counts for the pres
Reticulocyte Count appropriate data. Plot values and use the standard ence of nucleated RBCs.
Principle curve to determine hemoglobin values. 17. Describe the aspects of establishing a point-of-care
Absolute Reticulocyte 7. Describe the procedure for performing a testing program, including quality management and
Count microhematocrit. selection of instrumentation.
Corrected Reticulocyte 8. Identify sources of error in routine manual proce 18. Discuss the advantages and disadvantages of point-
Count dures discussed in this chapter and recognize written of-care testing as they apply to hematology tests.
Reticulocyte Production
scenarios describing such errors. 19. Describe the principles of common instruments
Index
9. Calculate the rule of three and erythrocyte indices used for point-of-care testing for hemoglobin level,
Reticulocyte Control
Flow Cytometry (mean cell volume, mean cell hemoglobin, and hematocrit, WBC counts, and platelet counts.
Erythrocyte mean cell hemoglobin concentration) when given
Sedimentation Rate appropriate data.
Principle
Modified Westergren
CASE STUDY
Erythrocyte
Sedimentation Rate After studying the material in this chapter, the reader should be able to respond to the following
Wintrobe Erythrocyte case studies:
Sedimentation Rate
Disposable Kits Case 1
Automated Erythrocyte The following results are obtained for a patient:
Sedimentation Rate RBCs4.63 1012/L
Point-of-Care Testing Hb15g/dL
Point-of-Care Tests Hct40% (0.40L/L)
Continued

172
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 173

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case studies:

1. Using the rule of three for hemoglobin/hematocrit, Case 3


what would be the expected value for the hemoglobin The following results are obtained for a patient using a point-
level? of-care device that employs the conductivity method to
2. What could cause the hemoglobin to be falsely elevated? measure the hematocrit:
3. What would you do to correct for the above interferences? Sodium160mmol/L (135 to 145)
Potassium3.6mmol/L (3.5 to 5.5)
Case 2 Hct17.0%
For another patient, the following results are obtained: Hb6.0g/dL
RBCs3.20 1012/L 1. Which electrolyte concentration could affect the
Hb5.8g/dL hematocrit?
Hct18.9% (0.189L/L) 2. Would this electrolyte concentration falsely decrease or
1. Calculate the RBC indices. increase the hematocrit value?
2. How would you describe the RBCs consistent with these 3. What other factors can decrease the hematocrit value?
indices?
3. How should you verify this?

A
s discussed in the introduction, clinical laboratory Equipment
hematology has evolved from simple observation and Hemacytometer
description of blood and its components to a highly The heart of the manual cell count is the hemacytometer, or
automated, extremely technical science including examination counting chamber. The most common one is the Levy chamber
at the molecular level. However, some of the more basic tests with improved Neubauer ruling. It is composed of two raised
have not changed dramatically over the years. This chapter surfaces, each in the shape of a 3-mm 3-mm square (total
provides an overview of these basic tests and presents the area 9mm2) separated by an H-shaped moat. As shown in
manual and semiautomated methods that can be used in lieu Figure 14-1, this large square is made up of nine 1-mm 1-mm
of automated instrumentation. Included in this chapter is a squares, and each of the white blood cell (WBC) squares is
discussion of point-of-care testing in hematology. divided further into 16 squares with the center square subdi-
vided into 25 smaller squares. Each of these smallest squares
is 1 25 or 0.04mm2. A coverslip is placed on top of the counting
surfaces. The distance between each counting surface and the
MANUAL CELL COUNTS
coverslip is 0.1mm; the total volume is 0.9mm3. Hemacytome
Although most routine cell-counting procedures in the hema- ters and coverslips must meet the specifications of the National
tology laboratory are automated, it may be necessary to use Bureau of Standards, as indicated by the initials NBS on the
manual methods when counts exceed the linearity of an instru- chamber. When the dimensions of the hemacytometer are
ment, when an instrument is nonfunctional and there is no thoroughly understood, the area counted can be changed to
backup, in remote laboratories in Third World countries, or in facilitate the counting of specimens with extremely low or high
a disaster situation when testing is done in the field. Although counts.
the discussion in this chapter concerns whole blood, body fluid
cell counts are also often performed using manual methods. Calculations
Chapter 17 discusses the specific diluents and dilutions used The general formula for manual cell counts is as follows and
for body fluid cell counts. Chapter 39 discusses automated can be used to calculate any type of cell count:
cell-counting instrumentation in detail.
cells counted dilution factor
Manual cell counts are performed using a hemacytometer, Total count =
area (mm 2 ) depth (0.1)
or counting chamber, and manual dilutions made with cali-
brated, automated pipettes and diluents (commercially avail- or
able or laboratory prepared). The principle for the performance
cells counted dilution factor 10*
of cell counts is essentially the same for leukocytes, erythro- Total count =
area (mm 2 )
cytes, and platelets; only the dilution, diluting fluid, and area
counted vary. Any particle (e.g., sperm) can be counted using
this system. *Reciprocal of depth.
174 PART III Routine Laboratory Evaluation of Blood Cells

1 mm

1 mm
R R

R R

side view

cover slip 0.1 mm


moat moat

Figure 14-1 Hemacytometer and a close-up view of the counting areas as seen under the microscope. The areas for the standard white blood cell count
are labeled W, and the areas for the standard red blood cell count are labeled R. The entire center square, outlined in blue, is used for counting platelets.

White Blood Cell Count


BOX 14-1 How to Make a Moist Chamber
The leukocyte or WBC count represents the number of WBCs
A moist chamber may be made by placing a piece of damp filter
in 1L of whole blood. Whole blood from a capillary puncture
paper in the bottom of a Petri dish. An applicator stick broken in half
or anticoagulated with ethylenediaminetetraacetic acid (EDTA)
can serve as a support for the chamber.
is mixed with a diluting fluid that contains a weak acid (acetic
or hydrochloric) or a buffered ammonium oxalate solution to
lyse the nonnucleated erythrocytes. The typical dilution for the
WBC count is 1:20. A hemacytometer is charged (filled) with
the well-mixed dilution and placed under a microscope to cells to give them time to settle. Care should be taken not
count the number of cells. to disturb the coverslip.
8. While keeping the hemacytometer in a horizontal posi-
PR O C E D U R E tion, place it on the microscope stage.
1. Clean the hemacytometer and coverslip with alcohol and 9. Lower the condenser on the microscope and focus by
dry thoroughly with a lint-free tissue. using the low-power (10) objective lens. The cells should
2. Make a 1:20 dilution, placing 25mcL of well-mixed be distributed evenly in all of the squares.
blood into 475mcL of weak acetic acid solution in a small 10. For a WBC 1:20 dilution, count all of the cells in the four
test tube. corner squares, starting with the square in the upper left-
3. Cover the tube and mix by inversion. hand corner (see Figure 14-1). Cells that touch the top and
4. Allow the dilution to sit for 10 minutes to ensure that the left lines should be counted; cells that touch the bottom
red blood cells (RBCs) have lysed. The solution will be and right lines should be ignored (Figure 14-2). See Figure
clear once lysis has occurred. Leukocyte counts should be 14-3 for the appearance of WBCs in the hemacytometer
performed within 3 hours of dilution. under the microscope.
5. Mix again by inversion and fill a plain microhematocrit 11. Repeat the count on the other side of the counting
tube. chamber. The difference between totals for the two sides
6. Charge both sides of the hemacytometer by holding the should be less than 10%. A greater variation could indicate
microhematocrit tube at a 45-degree angle and touching an uneven distribution, which requires that the procedure
the tip to the coverslip edge where it meets the chamber be repeated.
floor. 12. Average the two sides. Using the average, calculate the
7. After charging the counting chamber, place it in a moist WBC count using the first of the two equations given
chamber (Box 14-1) for 10 minutes before counting the earlier.
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 175

General reference ranges for males and females in different


age groups can be found on the inside front cover of this text.
Laboratory values may vary slightly according to the population tested
and should be established for each institution.

Sources of Error and Comments


1. The hemacytometer and coverslip should be cleaned
properly before they are used. Dust and fingerprints may
cause difficulty in distinguishing the cells.
2. The diluting fluid should be free of contaminants.
3. If the count is low, a greater area may be counted (e.g.,
9mm2) to improve accuracy.
4. The chamber must be charged properly to ensure an accu-
rate count. Uneven flow of the diluted blood into the
chamber results in an irregular distribution of cells. If the
chamber is overfilled or underfilled, the chamber must be
cleaned and recharged.
5. After the chamber is filled, allow the cells to settle for 10
minutes before counting.
6. Any nucleated erythrocytes (NRBCs) present in the sample
Figure 14-2 One square of a hemacytometer indicating which cells to
are not lysed by the diluting fluid. The NRBCs are counted
count. Cells touching the left and top lines (solid circles) are counted. Cells
as WBCs because they are indistinguishable when seen on
touching bottom and right (open circles) are not counted.
the hemacytometer. If five or more NRBCs per 100WBCs
are discovered on the differential, the WBC count must be
corrected for these cells. This is accomplished by using the
WBC
following formula:

Uncorrected WBC count 100


Number of NRBCs per 100 WBCs + 100

Report the result as the corrected WBC count.


7. The accuracy of the manual WBC count can be assessed by
performing a WBC estimate on the peripheral blood smear
(see Chapter 15). Boxes 14-2 and 14-3 show another
method of WBC estimation.

Platelet Count
A phase-contrast microscope is used in the reference method
for performing a manual platelet count. Platelets are adhesive
to foreign objects and to each other, which makes it difficult
Figure 14-3 White blood cells as seen in the hemacytometer under the to count them. They also are small and can be confused easily
10 objective. with dirt. In this procedure, whole blood, with EDTA as the
anticoagulant, is diluted with 1% ammonium oxalate, which
Example lyses the nonnucleated erythrocytes. The platelets can be
When a 1:20 dilution is used, the four large squares on one counted with the use of a phase-contrast microscope as
side of the chamber yield counts of 23, 26, 22, and 21. The described by Brecher and Cronkite.1
total count is 92. The four large squares on the other side of
the chamber yield counts of 28, 24, 22, and 26. The total count PROCEDURE
is 100. The difference between sides is less than 10%. 1. Make a 1:100 dilution, placing 20mcL of well-mixed blood
The average of the two sides of the chamber is 96. Using into 1980mcL of ammonium oxalate in a small test tube.
the average in the formula: 2. Mix the dilution thoroughly and charge the chamber.
(NOTE: A special thin flat-bottomed counting chamber is
cells counted dilution factor used for phase platelet counts.)
WBC count =
area counted (mm 2 ) depth 3. Place the charged hemacytometer in a moist chamber (see
96 20 Box 14-1) for 15 minutes to allow the platelets to settle.
=
4 0.1 4. Platelets are counted using the 40 objective lens. The plate-
= 4800 mm3 or 4.8 109 L lets have a diameter of 2 to 4m and appear round or oval,
176 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 14-1 Manual Cell Counts with Most Common


BOX 14-2 How to Determine the WBC Estimating Dilutions, Counting Areas
Factor
1. Perform automated WBC counts on 30 consecutive fresh patient Cell Diluting
blood samples. Make sure the WBC count is in control. Counted Fluid Dilution Objective Area Counted
2. Prepare and stain one peripheral blood film for each sample. Leukocyte Weak acid 1:20 10 or 40 4mm2
3. For each film, under oil-immersion microscopy, find an area where 1:100 10 9mm2
50% of the red cells are overlapping in doublets or triplets. Then Erythrocyte Isotonic 1:100 40 0.2mm2 (5 small
count the number of WBCs in 10 consecutive fields. saline squares of
4. Divide by 10 the total number of WBCs found to obtain the average center mm2)
number per single oil-immersion field. Platelet Ammonium 1:100 40 phase 1mm2
5. Divide the automated WBC count by the average number of WBCs oxalate
per oil field.
6. Add the numbers obtained in step 5 and divide by 30 (the number
of observations in this analysis) to obtain the average ratio of the
WBC count-to-WBCs per oil-immersion field. General reference ranges for males and females according
7. Round the number calculated in step 6 to the nearest whole number to age groups can be found on the inside front cover of
to obtain an estimation factor. this text.
Modified from Stiene-Martin EA: Clinical hematology: principles, procedures,
correlations, ed 2, Philadelphia, 1998, Lippincott. Sources of Error and Comments
1. Inadequate mixing and poor collection of the sample can
cause the platelets to clump on the hemacytometer. If the
problem persists after redilution, a new sample is needed.
A finger-stick sample is less desirable because of the adhe-
BOX 14-3 How to Perform a White Blood Cell (WBC) sive quality of the platelets.
Estimate 2. Dirt in the pipette, hemacytometer, or diluting fluid may
1. Scan a thin area, using the 50 oil immersion lens. cause the counts to be inaccurate.
2. Observe 10 fields, counting all WBCs in each field. 3. If fewer than 50 platelets are counted on each side, the
3. Average the number of WBCs seen per oil immersion field. procedure should be repeated by diluting the blood to 1:20
4. Multiply the number of WBCs per oil immersion field by 3000 and using calibrated pipettes. If more than 500 platelets are
compare with the WBC count. counted on each side, a 1:200 dilution should be made.
The appropriate dilution factor should be used in calculat-
From U.S. Army Medical Department Center and School Standard Operating
ing the results.
Procedure, Fort Sam Houston, Texas, November 1, 2001. Other methods of WBC
estimation are discussed in Chapter 15. 4. If the patient has a normal platelet count, the red cell area
(see Figure 14-1) may be counted. Then, the area is 0.2mm2
on each side.
5. The phenomenon of platelet satellitosis may occur when
EDTA anticoagulant is used. This refers to the adherence
displaying a light purple sheen when phase-contrast micros- of platelets around neutrophils, producing a ring or satel-
copy is used. The shape and color help distinguish the lite effect (see Figure 15-1). Using sodium citrate as the
platelets from highly refractile dirt and debris. Ghost anticoagulant should correct this problem. Because of
RBCs often are seen in the background. the dilution in the citrate tubes, it is necessary to multiply
5. Count the 25 small squares in the center square of the grid the obtained platelet count by 1.1 for accuracy (see
(see Figure 14-1). The area of this center square is 1mm2. Chapter 15).
Each side of the hemacytometer should be counted, and the
difference between the totals should be less than 10%.
6. Calculate the number of platelets per liter by using the first Erythrocyte Counts
equation given earlier. For example, if 200 platelets were Manual erythrocyte counts are rarely performed because of the
counted in the entire center square, inaccuracy of the count and questionable necessity. Use of
other, more accurate manual RBC parameters, such as the
200 100 microhematocrit and hemoglobin concentration, is desirable.
1 0.1 Table 14-1 contains information on performing manual eryth-
= 200, 000 mm3 or 200 109 L rocyte counts.

7. Platelet counts should be verified by performing an


estimate on the Wright-stained peripheral blood smear Body Fluid Cell Counts
(see Chapter 15). Body fluid cell counts are discussed in detail in Chapter 17.
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 177

HEMOGLOBIN DETERMINATION
The primary function of hemoglobin within the erythrocyte is
to carry oxygen to and carbon dioxide from the tissues. The
cyanmethemoglobin (hemoglobincyanide) method for hemo-
globin determination is the reference method approved by the
Clinical and Laboratory Standards Institute.2

Principle
In the cyanmethemoglobin method, blood is diluted in an
alkaline Drabkin solution of potassium ferricyanide, potassium
cyanide, sodium bicarbonate, and a surfactant. The hemoglo-
bin is oxidized to methemoglobin (Fe3+) by the potassium
ferricyanide, K3Fe(CN)6. The potassium cyanide (KCN) then
converts the methemoglobin to cyanmethemoglobin:

Hb ( Fe 2 + ) K 3Fe (CN )6 methemoglobin ( Fe 3+ ) KCN Figure 14-4 Spectrophotometer, used to measure transmittance or
cyanmethemoglobin absorbance.

The absorbance of the cyanmethemoglobin at 540nm is


directly proportional to the hemoglobin concentration. Sulf- be rinsed thoroughly with the reagent to ensure that no
hemoglobin is not converted to cyanmethemoglobin; it cannot blood remains. Follow the same procedure for the control
be measured by this method. Sulfhemoglobin fractions of samples.
more than 0.05g/dL are seldom encountered in clinical 4. Cover and mix well by inversion or using a vortex mixer.
practice, however.3 Let stand for 10 minutes at room temperature to allow full
conversion of hemoglobin to cyanmethemoglobin.
PROCEDURE 5. Transfer all of the solutions to cuvettes. Set the spectro
Hemoglobin Reagent photometer to 100% transmittance at the wavelength of
1. Create a standard curve, using a commercially available 540nm, using cyanmethemoglobin reagent as a blank.
cyanmethemoglobin standard. 6. Using a matched cuvette, continue reading the patient
a. When a standard containing 80mg/dL of hemoglobin samples and record the percentage transmittance.
is used, the following dilutions should be made: 7. Determine the hemoglobin values of the control samples
and the patient samples from the standard curve.
Hemoglobin General reference ranges can be found on the inside cover
Concentration (g/dL) Blank 5 10 15 20 of this text.
Cyanmethemoglobin 0 1.5 3 4.5 6
standard (mL) Sources of Error and Comments
Cyanmethemoglobin 6 4.5 3 1.5 0 1. Cyanmethemoglobin reagent is sensitive to light. It should
reagent (mL) be stored in a brown bottle or in a dark place.
2. A high leukocyte count (greater than 20 109/L) or a high
b. Transfer the dilutions to cuvettes. Set the wavelength platelet count (greater than 700 109/L) can cause turbidity
on the spectrophotometer (Figure 14-4) to 540nm and a falsely high result. In this case, the solution can be
and use the blank to set to 100% transmittance. centrifuged and the supernatant measured.
c. Using semilogarithmic paper, plot percentage 3. Lipemia also can interfere. A false result can be corrected by
transmittance on the y-axis and the hemoglobin adding 0.01mL of the patients plasma to 5mL of the
concentration on the x-axis. The hemoglobin cyanmethemoglobin reagent and using this solution as the
concentrations of the control and patient samples can reagent blank.
be read from this standard curve (Figure 14-5). 4. Cells containing Hb S and Hb C may be resistant to hemo-
d. A standard curve should be set up with each new lot of lysis, causing turbidity; this can be corrected by making a
reagents. It also should be checked when alterations 1:2 dilution with distilled water (1 part diluted sample plus
are made to the instrument (e.g., bulb change). 1 part water) and multiplying the results from the standard
2. Controls should be run with each batch of specimens. Com- curve by 2.
mercial controls are available. 5. Abnormal globulins, such as those found in patients with
3. Using the patients whole blood anticoagulated with EDTA multiple myeloma or Waldenstrm macroglobulinemia,
or heparin or blood from a capillary puncture, make a may precipitate. If this occurs, add 0.1g of potassium car-
1:251 dilution by adding 0.02mL (20mcL) of blood to bonate to the cyanmethemoglobin reagent. Commercially
5mL of cyanmethemoglobin reagent. The pipette should available cyanmethemoglobin reagent has been modified
178 PART III Routine Laboratory Evaluation of Blood Cells

10

15

20
% Transmittance

25

30

35

40

45

50

55

60
65

70
75

80
85

90
95
100
5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Hemoglobin concentration (g/dL)
Figure 14-5 Standard curve obtained when a cyanmethemoglobin standard of 80mg/dL is used. A blank (100% transmittance) and four dilutions were
made: 5g/dL (72.9% transmittance), 10g/dL (53.2% transmittance), 15g/dL (39.1% transmittance), and 20g/dL (28.7% transmittance).

to contain KH2PO4 salt, so this problem is not likely to 8. Commercial absorbance standards kits are available to cali-
occur. brate spectrophotometers.
6. Carboxyhemoglobin takes 1 hour to convert to cyanmethe- There also is a hand-held system in which hemoglobin is
moglobin and theoretically could cause erroneous results in converted to azide methemoglobin, which is read photometri-
samples from heavy smokers. The degree of error is proba- cally at two wavelengths (570nm and 880nm). This method
bly not clinically significant, however. avoids the necessity of sample dilution and interference from
7. Because hemoglobin reagent contains cyanide, it must be turbidity. An example is the HemoCue (HemoCue, Inc., Lake
used cautiously; a minimum of 4L of reagent is lethal. Acid- Forest, Calif),4,5 which is also discussed later in the section on
free sinks should be used for disposal of reagent and point-of-care testing. Another method that has been used in
samples, because acidification of cyanide releases hydrogen some automated instruments involves the use of sodium lauryl
cyanide gas. Copious amounts of water should be used to sulfate (SLS) to convert hemoglobin to SLS-methemoglobin.
flush the sink after disposal. This method does not generate toxic wastes.6-9
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 179

MICROHEMATOCRIT
The hematocrit is the volume of packed RBCs that occupies a
given volume of whole blood. This is often referred to as the
packed cell volume (PCV). It is reported either as a percentage
(e.g., 36%) or in liters per liter (0.36L/L).

PROCEDURE
1. Fill two plain capillary tubes approximately three quarters
full with blood anticoagulated with EDTA or heparin. Mylar-
wrapped tubes are recommended by the National Institute
for Occupational Safety and Health to reduce the risk of
capillary tube injuries.10 Alternatively, blood for heparinized
capillary tubes may be collected by capillary puncture. Wipe
any excess blood from the outside of the tube.
2. Seal the end of the tube with the colored ring using nonab-
sorbent clay. Hold the filled tube horizontally and seal by
placing the dry end into the tray with sealing compound at
a 90-degree angle. Rotate the tube slightly and remove it
from the tray. The plug should be at least 4mm long.10 Figure 14-6 Microhematocrit reader.
3. Balance the tubes in the centrifuge with the clay ends facing
the outside away from the center, touching the rubber
gasket.
4. Tighten the head cover on the centrifuge and close the top.
Centrifuge the tubes at between 10,000g and 15,000g for
the time that has been determined to obtain maximum
packing of RBCs, as detailed in Box 14-4. Do not use the
brake to stop the centrifuge.
5. Determine the hematocrit by using a microhematocrit
reading device (Figure 14-6). Read the level of RBC packing;
do not include the buffy coat (leukocytes and platelets)
when reading (Figure 14-7).
6. The values of the duplicate hematocrits should agree within
1% (0.01L/L).10
General reference ranges according to sex and age can be
found on the inside front cover of this text.
Plasma
Sources of Error and Comments
1. Improper sealing of the capillary tube causes a decreased
hematocrit reading as a result of loss of blood during
centrifugation. A higher number of erythrocytes are lost
compared with plasma.

BOX 14-4 Determining Maximum Packing Time for


Microhematocrit Buffy coat
The time to obtain maximum packing of red blood cells should be (white blood cells
and platelets)
determined for each centrifuge. Duplicate microhematocrit determi
nations should be made using fresh, well-mixed blood anticoagu Red blood cells
lated with ethylenediaminetetraacetic acid. Two specimens should
be used, with one of the specimens having a known hematocrit of
50% or higher. Starting at 2 minutes, spin duplicates at 30-second
intervals and record results. When the hematocrit has remained at Clay
the same value for two consecutive readings, optimum packing has
been achieved, and the second time interval should be used for Figure 14-7 Capillary tube after it has been centrifuged. Notice the layers
microhematocrit determinations.10 containing plasma, the buffy coat (leukocytes and platelets), and the
erythrocytes.
180 PART III Routine Laboratory Evaluation of Blood Cells

2. An increased concentration of anticoagulant decreases the


hematocrit reading as a result of erythrocyte shrinkage.
3. A decreased or increased result may occur if the specimen
was not mixed properly.
4. The time and speed of the centrifugation and the time
when the results are read are important. Insufficient cen-
trifugation or a delay in reading results after centrifugation
causes hematocrit readings to increase. Time for complete
packing should be determined for each centrifuge and
rechecked at regular intervals. When the microhematocrit
centrifuge is calibrated, one of the specimens used must
have a hematocrit of 50% or higher.10
5. The buffy coat of the specimen should not be included in
the hematocrit reading because this falsely elevates the
result.
6. A decrease or increase in the readings may be seen if the
microhematocrit reader is not used properly.
7. Many disorders, such as sickle cell anemia, macrocytic
anemias, hypochromic anemias, spherocytosis, and thalas-
semia, may cause plasma to be trapped in the erythrocytes
layer even if the procedure is performed properly. The
trapping of the plasma causes the microhematocrit to be
Figure 14-8 READACRIT centrifuge with built-in capillary tube compart
1% to 3% (0.01 to 0.03L/L) higher than the value obtained
ments and hematocrit scales. (Courtesy and Becton, Dickinson and
using automated instruments that calculate the hematocrit Company, Franklin Lakes, N.J.)
and are unaffected by the trapped plasma.
8. A temporarily low hematocrit reading may result immedi-
ately after a blood loss because plasma is replaced faster Case 1
than are erythrocytes. Hb = 12g/dL
9. The fluid loss associated with dehydration causes a decrease Hct = 36% (0.36L/L)
in plasma volume and falsely increases the hematocrit According to the rule of three,
reading.
10. Proper specimen collection is an important consideration. Hb (12) 3 = Hct (36)
The introduction of interstitial fluid from a skin puncture
or the improper flushing of a catheter causes decreased An acceptable range for the hematocrit would be 33% to
hematocrit readings. 39%. These values conform to the rule of three.
The READACRIT centrifuge (Becton, Dickinson and
Company, Franklin Lakes, N.J.) uses precalibrated capillary Case 2
tubes and has built-in hematocrit scales, which eliminates the Hb = 9g/dL
need for separate reading devices (Figure 14-8). The use of Hct = 32%
SUREPREP Capillary Tubes (Becton Dickinson) eliminates the According to the rule of three,
use of sealants. They have a factory-inserted plug that seals
automatically when the blood touches the plug.11 Hb (9.0) 3 = Hct (27 versus actual value of 32)

An acceptable range for hematocrit would be 24% to 30%,


RULE OF THREE
so these values do not conform to the rule of three.
When specimens are analyzed by automated or manual
methods, a quick visual check of the results of the hemoglobin Case 3
and hematocrit can be done by applying the rule of three. Hb = 15g/dL
This rule applies only to specimens that have normocytic nor- Hct = 36%
mochromic erythrocytes. The value of the hematocrit should According to the rule of three,
be three times the value of the hemoglobin plus or minus 3:
Hb 3 = Hct 3 (0.03L/L). It should become habit for the Hb (15) 3 = Hct (45 versus obtained value of 36)
analyst to multiply the hemoglobin by 3 mentally for every
specimen; a value discrepant with this rule may indicate abnor- An acceptable range for hematocrit would be 42% to 48%,
mal RBCs, or it may be the first indication of error. so these values do not conform to the rule of three.
For example, the following results are obtained from If values do not agree, the blood smear should be examined
patients: for abnormal erythrocytes. In the second example, the blood
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 181

TABLE 14-2 Red Blood Cell Indices and Correlating Red Blood Cell Morphology and Disease States
MCV (fL) MCHC (g/dL) Red Blood Cell Morphology Found in
<80 <32 Microcytic; hypochromic Iron deficiency anemia, thalassemia, sideroblastic anemia, other conditions of
defective iron use, chronic infection or inflammation, unstable hemoglobins
80-100 32-36 Normocytic; normochromic Hemolytic anemia, leukemia, metastatic malignancy, bone marrow failure,
chronic renal disease
>100 32-36 Macrocytic; normochromic Liver disease, myelodysplasias, megaloblastic anemias

MCHC, Mean cell hemoglobin concentration; MCV, mean cell volume.

film reveals erythrocytes that are abnormal in hemoglobin For example, if the hemoglobin = 16g/dL and the RBC count
content (hypochromic), so the values obtained are accurate. If = 5 1012/L, MCH = 32pg.
erythrocytes do appear normal, the problem should be inves- The reference range for adults is 28 to 32pg. The MCH
tigated. In the third example, the specimen is determined to generally is not considered in the classification of anemias.
have lipemic plasma, and a correction must be made to the
hemoglobin level to obtain an accurate hemoglobin value. Mean Cell Hemoglobin Concentration
(See Hemoglobin Determination in this chapter.) The MCHC is the average concentration of hemoglobin in each
When an unexplained discrepancy is found, the specimen individual erythrocyte. The units used are grams per deciliter
processed before and after the specimen in question should be (formerly given as a percentage):
checked to determine whether they conform to the rule. If they
do not conform, further investigation should be done to find MCHC = Hb ( g dL ) 100 Hct (% )
the problem. A control specimen should be run when such a
discrepancy is found. If the instrument produces appropriate For example, if the Hb = 16g/dL and the Hct = 48%, MCHC
results for the control, random error may have occurred (see = 33.3g/dL.
Chapter 5). Values of normochromic cells range from 32 to 36g/dL;
values of hypochromic cells are less than 32g/dL, and values
of hyperchromic cells are greater than 36g/dL. Hypochromic
RED BLOOD CELL INDICES
erythrocytes occur in thalassemias and iron deficiency. The
The mean cell volume (MCV), mean cell hemoglobin (MCH), term hyperchromic is a misnomer: a cell does not really contain
and mean cell hemoglobin concentration (MCHC) are RBC more than 36g/dL of hemoglobin, but its shape may have
indices. These are calculated to determine the size and hemo- become spherocytic, which makes the cell appear full. An
globin content of the average RBC. In addition to serving as a MCHC greater than 36g/dL should be scrutinized carefully
quality control check, the indices may be used to differentiate for an error in hemoglobin value (see Sources of Error and
anemias. Table 14-2 provides a summary of the RBC indices, Comments in the section on hemoglobin determination).
morphology, and correlation with anemias. The morphologic Another cause for a markedly increased MCHC could be the
classification of anemia on the basis of MCV is discussed in presence of a cold agglutinin. Incubating the specimen at 37
detail in Chapter 18. C for 15 minutes before analysis usually produces accurate
results. Cold agglutinin disease is discussed in more detail in
Mean Cell Volume Chapter 25.
The MCV is the average volume of the RBC, expressed in fem-
toliters (fL), or 1015L:
RETICULOCYTE COUNT
MCV = Hct (% ) 10 RBC count ( 10 12
L) The reticulocyte is the last immature erythrocyte stage. Nor-
mally, a reticulocyte spends 2 to 3 days in the bone marrow
For example, if the Hct = 45% and the RBC count = 5 1012/L, and 1 day in the peripheral blood before developing into a
MCV = 90fL. mature erythrocyte. The reticulocyte contains remnant cyto-
The reference range for MCV is 80 to 100fL. RBCs with an plasmic ribonucleic acid (RNA) and organelles such as the
MCV of less than 80fL are microcytic; those with an MCV of mitochondria and ribosomes (see Chapter 8). The reticulocyte
more than 100fL are macrocytic. count is used to assess the erythropoietic activity of the bone
marrow.
Mean Cell Hemoglobin
The MCH is the average weight of hemoglobin in an RBC, Principle
expressed in picograms (pg), or 1012g: Whole blood, anticoagulated with EDTA, is stained with a
supravital stain, such as new methylene blue. Any nonnucle-
MCH = Hb ( g dL ) 10 RBC count ( 1012 L ) ated erythrocyte that contains two or more particles of
182 PART III Routine Laboratory Evaluation of Blood Cells

Figure 14-9 Reticulocytes under an oil immersion lens (peripheral blood


1000).

blue-stained granulofilamentous material after new methylene


blue staining is defined as a reticulocyte (Figure 14-9).
Figure 14-10 Miller ocular disc. Alternatively, square B may be in the
center of square A.
PR O C E D U R E
1. Mix equal amounts of blood and new methylene blue stain
(2 to 3 drops, or 50mcL each) and allow to incubate at the College of American Pathologists (CAP) hematology
room temperature for 3 to 10 minutes.12 standard for a manual reticulocyte count based on at least
2. Remix the preparation. 1000 red cells.13 The calculation formula for percent reticulo-
3. Prepare two wedge films (see Chapter 15). cytes is
4. In an area in which cells are close together but not touching,
count 1000RBCs under the oil immersion objective lens. No. reticulocytes in square A
Reticulocytes are included in the total RBC count (i.e., a (large square ) 100
Reticulocytes % =
reticulocyte counts as both an RBC and a reticulocyte). No. RBCs in square B (small square ) 9
5. To improve accuracy, have another laboratorian count the
other smear; values should agree within 20%. Reference Range
6. Calculate the reticulocyte count: General reference ranges can be found on the inside front cover
of this text.
Number of reticulocytes 1000 (RBCs observed ) 100 =
percentage reticulocytes Sources of Error and Comments
1. If a patient is very anemic or polycythemic, the proportion
For example, if 15 reticulocytes are counted, of dye to blood should be adjusted accordingly.
2. An error may occur if the blood and stain are not mixed
Reticulocyte (% ) = 15 1000 100 = 1.5% before the smears are made. The specific gravity of the
reticulocytes is lower than that of the mature erythrocytes,
Or the number of reticulocytes counted can be multiplied by and reticulocytes settle at the top of the mixture during
0.1 (100/1000) to obtain the result. incubation.
3. Moisture in the air, poor drying of the slide, or both may
Miller Disc cause areas of the slide to appear refractile, and these areas
Because large numbers of red cells should be counted to obtain could be confused with reticulocytes. The RNA remnants in
a more precise absolute reticulocyte count, the Miller disc was a reticulocyte are not refractile.
designed to reduce this labor-intensive process. The disc is 4. Other RBC inclusions that stain supravitally include Heinz,
composed of two squares, with the area of the smaller square Howell-Jolly, and Pappenheimer bodies. Heinz bodies rep-
measuring 1 9 the area of the larger square. The disc is inserted resent precipitated hemoglobin, usually appear round or
into the eyepiece of the microscope and the grid in Figure oval, and tend to cling to the cell membrane (Figure 14-11).
14-10 is seen. RBCs are counted in the smaller square and Howell-Jolly bodies are round nuclear fragments and are
reticulocytes are counted in the larger square. Selection of the usually singular. Pappenheimer bodies are hemosiderin in
counting area is the same as described earlier. A minimum of the mitochondria whose presence can be confirmed with an
112 cells should be counted in the small square, because this iron stain, such as Prussian blue. This stain is discussed in
is equivalent to 1008 red cells in the large square and satisfies Chapter 16.
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 183

to compensate for anemia. The corrected reticulocyte count


depends on the degree of anemia.
B
Reticulocyte Production Index
Principle
Reticulocytes that are released from the marrow prematurely
B are called shift reticulocytes. These reticulocytes are shifted from
the bone marrow to the peripheral blood earlier than usual to
compensate for anemia. Instead of losing their reticulum in 1
A day, as do normal reticulocytes, these cells take 2 to 4 days to
lose their reticula. When erythropoiesis is evaluated, a correc-
tion should be made for the presence of shift reticulocytes if
A there is a report of polychromasia in the morphology. Normal
(nonshift) reticulocytes become mature RBCs 1 day after enter-
ing the bloodstream and thus represent 1 days production of
RBCs in the bone marrow. Cells shifted to the peripheral blood
prematurely stay longer as reticulocytes and contribute to the
Figure 14-11 Reticulocytes (A) and Heinz bodies (B) stained with supra reticulocyte count for more than 1 day. For this reason, the
vital stain (peripheral blood 1000). reticulocyte count is falsely increased when polychromasia is
present, because the count no longer represents the cells matur-
ing in just 1 day. On many automated instruments, this math-
Absolute Reticulocyte Count ematical adjustment of the reticulocyte count has been replaced
Principle by the measurement of immature reticulocyte fraction.12
The absolute reticulocyte count (ARC) is the actual number of The patients hematocrit is used to determine the appropri-
reticulocytes in 1L of whole blood. ate correction factor (reticulocyte maturation time in days):

Calculations Patients Hematocrit Correction Factor


Value (%) (Maturation) (days)
reticulocytes (% ) RBC count ( 1012 L ) 40-45 1
ARC =
100 35-39 1.5
25-34 2
For example, if a patients reticulocyte count is 2% and the RBC 15-24 2.5
count is 2.20 1012/L, the ARC would be calculated as follows: <15 3

2 ( 2.20 1012 /L )
ARC = = 44 109 L Calculation
100 The reticulocyte production index (RPI) is calculated as follows:

Reference Range reticulocyte (% ) [Hct (% ) 45]


RPI =
Values between 25 10 /L and 75 10 /L are within the refer-
9 9 maturation time
ence range for most populations.14
or

Corrected Reticulocyte Count corrected reticulocyte count


Principle RPI =
maturation time
In specimens with a low hematocrit, the percentage of reticu-
locytes may be falsely elevated because whole blood contains For example, for a patient with a reticulocyte count of 7.8%
fewer RBCs. A correction factor is used, with the average normal and Hct of 30%, and with polychromasia noted, the previous
hematocrit considered to be 45%. table indicates a maturation time of 2 days. Thus

Calculation 7.8 [30 45]


RPI =
2
patient Hct
Corrected reticulocyte count = reticulocytes (% )
5
45 RPI = 2.6

Reference Range Reference Range


Patients with a hematocrit of 35% are expected to have a cor- An adequate bone marrow response usually is indicated by
rected reticulocyte count of 2% to 3%. In patients with a hema- an RPI that is greater than 3. An inadequate erythropoietic
tocrit of less than 25%, the count should increase to 3% to 5% response is seen when the RPI is less than 2.14
184 PART III Routine Laboratory Evaluation of Blood Cells

Reticulocyte Control Normal erythrocytes have a relatively small mass and settle
Several commercial controls are now available for monitoring slowly. Certain diseases can cause rouleaux formation, in
manual and automated reticulocyte counts [e.g., Retic-Chex II, which the plasma fibrinogen and globulins are altered. This
Streck Laboratories, Omaha, Nebr.; Liquichek Reticulocyte alteration changes the erythrocyte surface, which leads to
Control (A), Bio-Rad Laboratories, Hercules, Calif.]. Most of aggregation of the RBCs, increased RBC mass, and a more
the controls are available at three levels. The control samples rapid ESR. The ESR is directly proportional to the RBC
are treated in the same manner as the clinical specimens. The mass and inversely proportional to plasma viscosity. Several
control can be used to verify the laboratorians accuracy and methods, both manual and automated, are available for mea-
precision when manual counts are performed. suring the ESR. Only the most commonly used methods are
discussed here.
Flow Cytometry
Most of the major instrument manufacturers offer analyzers Modified Westergren Erythrocyte
that perform automated reticulocyte counts. All of the analyzers Sedimentation Rate
evaluate reticulocytes based on optical scatter or fluorescence The most commonly used method today is the modified
after the RBCs are treated with fluorescent dyes or nucleic acid Westergren method. One advantage of this method is that the
stains to stain residual RNA in the reticulocytes. The percentage taller column height allows the detection of highly elevated
and the absolute count are provided. These results are statisti- ESRs. It is the method recommended by the International
cally more valid because of the large number of cells counted. Council for Standardization in Hematology and the Clinical
Other reticulocyte parameters that are offered on some auto- and Laboratory Standards Institute.18,19
mated instruments include maturation index, immature reticu-
locyte fraction, and reticulocyte indices. These values may be PROCEDURE
especially useful in monitoring erythropoietic activity in 1. Use blood collected in EDTA and dilute at four parts blood
patients undergoing chemotherapy or stem cell or bone marrow to one part 3.8% sodium citrate or 0.85% sodium chloride
transplantation or in patients with chronic renal failure. Auto- (e.g., 2mL blood and 0.5mL diluent). Alternatively, blood
mated reticulocyte counting is discussed in Chapter 39. can be collected directly into special sedimentation test
tubes containing sodium citrate. Standard coagulation test
tubes are not acceptable, because the dilution is nine parts
ERYTHROCYTE SEDIMENTATION RATE
blood to one part sodium citrate.19
The erythrocyte sedimentation rate (ESR) is commonly used as 2. Place the diluted specimen in a 200-mm column with an
a general screening test. An elevated ESR in the absence of internal diameter of 2.55mm or more.
specific symptoms may guide the clinician to further evalua- 3. Place the pipette into the rack and allow to stand undis-
tion. The ESR is useful for monitoring the course of an existing turbed for 60 minutes.
inflammatory disease or for differentiating between similar 4. Record the number of millimeters the RBCs have fallen. The
diseases. For example, the ESR is normal in patients with osteo- buffy coat should not be included in the reading. The ESR
arthritis but is elevated in patients with rheumatic fever, rheu- is reported as 1 hour = __ mm.
matoid arthritis, or pyogenic arthritis. The ESR is elevated in
the early stage of acute pelvic inflammatory disease or a rup- Wintrobe Erythrocyte Sedimentation Rate
tured ectopic pregnancy but normal in the first 24 hours of When the Wintrobe method (Figure 14-12) was first intro-
acute appendicitis. The ESR may be used to assess the activity duced, the sample used was oxalate-anticoagulated whole
of pulmonary tuberculosis. More recent studies have examined blood. This was placed in a 100-mm column. Today, EDTA-
the use of the C-reactive protein level as a more applicable, treated or citrated whole blood is used with the shorter column,
predictable, and responsive indicator of early postoperative and the name implies only that the shorter column was used.
infectious complications.15 The shorter column height allows a somewhat increased sen-
sitivity in detecting mildly elevated ESRS.
Principle
When anticoagulated blood is allowed to stand at room tem- PROCEDURE
perature undisturbed for a period of time, the RBCs settle 1. Use fresh blood collected in EDTA anticoagulant. A
toward the bottom of the tube. The ESR is the distance in mil- minimum of 2mL of whole blood is needed.
limeters that the RBCs fall in 1 hour. The ESR is affected by 2. After mixing the blood thoroughly, fill a Pasteur pipette
erythrocytes, plasma, and mechanical and technical factors. using a rubber pipette bulb.
Erythrocytes have a net negative surface charge and tend to 3. Place the filled pipette into the Wintrobe tube until the tip
repel one another. The repulsive forces are partially or totally reaches the bottom of the tube.
counteracted if there are increased quantities of positively 4. Carefully squeeze the bulb and expel the blood into the
charged plasma proteins; the erythrocytes settle more rapidly Wintrobe tube while pulling the Pasteur pipette up from the
as a result of the formation of RBC aggregates or rouleaux. bottom of the tube. There must be steady, even pressure
Examples of macromolecules that can produce this reaction are on the bulb and expulsion of blood into the tube as well
fibrinogen, -globulins, and pathologic immunoglobulins.16,17 as continuous movement of the pipette up the tube to
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 185

Figure 14-12 Wintrobe sedimentation rate system. (Courtesy Lynda


Meyer, MT(ASCP), Memphis, Tenn.)

prevent the introduction of air bubbles into the column of


blood.
5. Fill the Wintrobe tube to the 0 mark.
6. Place the tube into a Wintrobe rack (tube holder) and allow
to stand undisturbed for 1 hour at room temperature. The Figure 14-13 Sediplast (Polymedco) disposable sedimentation rate
rack must be perfectly level and placed in a draft-free room. system. (Courtesy Polymedco, Cortlandt Manor, N.Y.)
7. Record the number of millimeters the RBCs have fallen.
Read the tube from the bottom of the plasma meniscus to
the top of the sedimented red cells. The result is reported 10. The ESR of patients with severe anemia is of little diagnos-
in millimeters per hour. tic value, because it will be falsely elevated.

Reference Range Disposable Kits


Reference ranges according to sex and age can be found on the Disposable commercial kits are available for ESR testing (Figure
inside front cover of this text. Table 14-3 lists some of the 14-13). Several kits include safety caps for the pipettes that
factors that influence the ESR. allow the blood to fill precisely to the zero mark. This safety
cap makes the pipette a closed system and eliminates the error
Sources of Error and Comments involved in manually setting the blood to the zero mark.
1. If the concentration of anticoagulant is increased, the ESR
is falsely low as a result of sphering of the RBCS, which Automated Erythrocyte Sedimentation Rate
inhibits rouleaux formation. There are several automated ESR systems available using the
2. The anticoagulants sodium or potassium oxalate and traditional Westergren and Wintrobe methods as well as alter-
heparin cause the RBCs to shrink and falsely elevate nate methods such as centrifugation. The Ves-Matic system
the ESR. (Diesse, Inc., Hialeah, Fla.) is a bench-top analyzer designed
3. A significant change in the temperature of the room alters to determine ESR by use of an optoelectronic sensor, which
the ESR. measures the change in opacity of a column of blood as sedi-
4. Even a slight tilt of the pipette causes the ESR to increase. mentation of blood progresses. Blood is collected in special
5. If the specimen is allowed to sit at room temperature for Ves-Tec or Vacu-Tec tubes, which contain sodium citrate and
more than 2 hours before being tested, the erythrocytes are compatible with the Vacutainer system. These tubes are used
start to become spherical, which may inhibit the forma- directly in the instrument (Figure 14-14). Acceleration of sedi-
tion of rouleaux. mentation is achieved by positioning the tubes at an 18-degree
6. Bubbles in the column of blood invalidate the test results. angle in relation to the vertical axis. Results comparable with
7. The blood must be filled properly to the zero mark at the Westergren 1-hour values are obtained in 20 minutes.20
beginning of the test. Another automated ESR analyzer is the Sedimat 15
8. A clotted specimen cannot be used. (Polymedco, Cortlandt Manor, N.Y.), which uses the principle
9. Hematologic disorders that prevent the formation of rou- of infrared measurement. It is capable of testing one to eight
leaux (e.g., the presence of sickle cells and spherocytes) specimens randomly or simultaneously and provides results in
decrease the ESR. 15 minutes (Figure 14-15).
186 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 14-3 Factors Affecting the Erythrocyte Sedimentation Rate (ESR)


Category Increased ESR Decreased ESR
Blood proteins and lipids Hypercholesterolemia Hyperalbuminemia
Hyperfibrinogenemia Hyperglycemia
Hypergammaglobulinemia Hypofibrinogenemia
Hypoalbuminemia Hypogammaglobulinemia
Increased bile salts
Increased phospholipids
Red blood cells Anemia Acanthocytosis
Macrocytosis Anisocytosis (marked)
Hemoglobin C
Microcytosis
Polycythemia
Sickle cells
Spherocytosis
Thalassemia
White blood cells Leukemia Leukocytosis (marked)
Drugs Dextran Adrenocorticotropic hormone (corticotropin)
Heparin Cortisone
Penicillamine Ethambutol
Procainamide Quinine
Theophylline Salicylates
Vitamin A
Clinical conditions Acute heavy metal poisoning Cachexia
Acute bacterial infections Congestive heart failure
Collagen vascular diseases Newborn status
Diabetes mellitus
End-stage renal failure
Gout
Malignancy
Menstruation
Multiple myeloma
Myocardial infarction
Pregnancy
Rheumatic fever
Rheumatoid arthritis
Syphilis
Temporal arteritis
Specimen handling Refrigerated sample not returned to room temperature Clotted blood sample
Delay in testing
Technique High room temperature Bubbles in ESR column
Tilted ESR tube Low room temperature
Vibration Narrow ESR column diameter

From American Society for Clinical Pathology/American Proficiency Institute: 2006 2nd Test EventEducational CommentaryThe Erythrocyte Sedimentation Rate and Its Clinical
Utility. API is the proficiency testing group that provides testing materials to the American Society for Clinical Pathology. The educational commentary itself is written by ASCP.
This reference can also be accessed at: http://www.api-pt.com/Reference/Commentary/2006Bcoag.pdf. Accessed June 24, 2010.

The ESR STAT PLUS system (HemaTechnologies, Lebanon,


POINT-OF-CARE TESTING
N.J.) is based on centrifugation. The advantages of this method
are a smaller required sample volume and shorter testing time, Point-of-care testing offers the ability to produce rapid and
which makes it more suitable for a pediatric patient popula- accurate results that help facilitate faster treatment, which can
tion. The disadvantage of this method is the number of exact- decrease patient length of stay. This testing is rarely performed
ing preanalytical steps that must be strictly followed to prevent by trained laboratory personnel; most often, it is carried out
erroneous results. Compliance with these steps may be difficult by nurses. Manufacturers have created analyzers with nonlabo-
to achieve consistently in a busy hematology laboratory.21 ratory operators in mind, but results obtained using these
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 187

A B
Figure 14-14 Two models of the Ves-Matic instruments for sedimentation rates: The Ves-Matic Easy (A) for up to 10 samples (requiring special tubes) and
the Ves-Matic Cube 30 (B) for up to 30 samples, determining the ESR directly from EDTA tubes. Products with up to 190 sample capacity are also available.
(Courtesy Diesse Inc., Hialeah, Fla.)

waived or moderately complex. Tests are classified as waived


if they are determined to be simple tests with an insignificant
risk of an erroneous result. Point-of-care testing is commonly
performed in hospital inpatient units, outpatient clinics, surgery
centers, emergency departments, long-term care facilities, and
dialysis units. For waived point-of-care testing, facilities are
required to obtain a certificate of waiver, pay the appropriate
fees, and follow the manufacturers testing instructions.22 For
any point-of-care program to be successful, certain key elements
must be present. Clear administrative responsibility, well-written
procedures, a training program, quality control, proficiency
testing, and equipment maintenance are essential for success.
The first step is appointing a laboratory point-of-care testing
coordinator. This person not only is the go-to person but is
also an important liaison between the laboratory and nursing
staff. The second step to ensuring a successful program is to create
a multidisciplinary team with authority to impact all aspects of
the POC program. This committee would have the authority to
oversee the integrity and quality of the existing POC program
and institute changes or new testing as needed. It is also impor-
Figure 14-15 Sedimat 15 (Polymedco) automated sedimentation rate tant to have administrative support to help remove barriers.
system. (Courtesy Polymedco, Cortlandt Manor, N.Y.) A point-of-care testing program must incorporate all of the
following. A written policy should be developed that defines
systems are still affected by preanalytic and analytic variables. the program. This policy should outline who is responsible for
The laboratorys partnership with nursing is the key to success each part of the program. The policy should also indicate
in any hospitals point-of-care program. where the testing is to be performed and who is going to
Point-of-care testing is defined as diagnostic testing at or near perform the testing. Testing procedures should be written that
the site of patient care. The Clinical Laboratory Improvement clearly state how to perform the tests and that address how to
Amendments of 1988 (CLIA) introduced the concept of testing handle critical values and/or any discrepant results. The
site neutrality, which means that regardless of where the diag- program must be monitored. An ongoing evaluation of the
nostic testing is performed or who performs the test, all testing point-of-care testing is vital for success.
sites must follow the same regulatory requirements based on the When the instrument to be used in the point-of-care testing
complexity of the test. Under CLIA, point-of-care testing program is being selected, it is helpful to invite the vendors to
(including physician-performed microscopy) is classified as demonstrate their equipment. An equipment display that is
188 PART III Routine Laboratory Evaluation of Blood Cells

available for hands-on use by the operators can be very helpful


in selection of the appropriate instrumentation. Patient corre-
lation studies are very useful in choosing equipment that best
covers the patient population for that particular institution.
Point-of-care operators need hand-held analyzers that are light-
weight, accurate, fast, and require little specimen material. The
point-of-care testing system should also address the following
laboratory concerns:
What is the range of measurement?
How well does the test system correlate with laboratory
instrumentation?
Can it be interfaced to the laboratory information system?
Does it give reliable results?
Does the company supply excellent technical support?
Is it affordable?
Paramount to point-of-care testing is patient safety. It is
important to maintain good practices, and with waived testing,
this often comes down to the basics. Such basics include proper
and appropriate specimen collection, proper identification of
the patient and specimen, proper storage of reagents, and good
documentation of patient test results (use of point-of-care
interfaces is beneficial), as well as proper performance of any
necessary instrument maintenance. Laboratory oversight is
Figure 14-16 i-STAT instrument for measuring hematocrit. (Courtesy
sometimes absent, and basic safety precautions necessary for Abbott Laboratories, Abbott Park, Ill.)
waived tests can be easily overlooked, often due to a lack of
understanding, lack of training, and high personnel turnover
rates.23 Patient safety, risk management, and error reduction
are primary goals of all healthcare facilities. All testing person-
nel should be properly trained in best practices to avoid
exposure. The individual responsible for oversightwhether
laboratory or nonlaboratorymust avoid taking safety for
granted. All applicable standards (including those of the Occu-
pation Safety and Health Administration, Centers for Disease
Control and Prevention, Joint Commission, CAP, CLIA, and so
forth) should be implemented and easily accessible. Because
the number of waived tests has grown significantly since waived
tests were first defined by CLIA, it is paramount that standard
safety precautions and the basic steps outlined earlier be imple-
mented to ensure that patient safety is not sacrificed in the
unique situation of CLIA-waived testing.

Point-of-Care Tests
Various point-of-care instruments are available to measure
parameters such as hemoglobin level and hematocrit, and
some perform a complete blood count.

Hematocrit Figure 14-17 Epoc device for measuring hematocrit. (Courtesy Epocal,
The most common methods for determining the hematocrit Inc., Ottawa, Ont., Canada.)
include the microhematocrit centrifuge, conductometric
methods, and calculation by automated cell counters (see comments in the Microhematocrit section). Examples of
Chapter 39). centrifuge-based devices are the Hematastat II (Separation
Centrifuge-based microhematocrit systems have been Technology, Inc., Altamonte Springs, Fla.) and STAT Crit
around for years, and the results obtained correlate well with (Wampole Laboratories, Cranbury, N.J.)
the results produced by standard cell counters. Nonlaboratori- The i-STAT 1 (Abbott Laboratories, Abbott Park, Ill.)24
ans and inexperienced operators, however, may be unaware of (Figure 14-16) as well as the Epoc (Epocal, Inc., Ottawa, Ont.)
the error that can be introduced by insufficient centrifugation (Figure 14-17)25 use the conductivity method to determine the
time and inaccurate reading of the microhematocrit tube (see hematocrit. Plasma conducts electrical current, whereas WBCs
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 189

act as insulators. In the i-STAT system, before the measured


sample conductance is converted into the hematocrit value,
corrections are applied for the temperature of the sample, the
size of the fluid segment being measured, and the relative
conductivity of the plasma component. The first two correc-
tions are determined from the measured value of the calibrant
conductance and the last correction from the measured con-
centrations of sodium and potassium in the sample.24

Sources of Error and Comments. Conductivity of a


whole blood sample is dependent upon the amount of
electrolytes in the plasma portion. Conductivity does not dis-
tinguish RBCs from other nonconductive elements such as
proteins, lipids, and WBCs that may be present in the sample.
A low total protein level will falsely decrease the hematocrit. Figure 14-18 Gem Premier instrument for measuring hematocrit.
The presence of lipids can interfere with the hematocrit. An (Courtesy Instrumentation Laboratory Company, Lexington, Mass.)
increased WBC count will falsely increase the hematocrit.
The presence of cold agglutinins can falsely decrease the spectrophotometric method. The STAT-Site MHbg Meter
hematocrit.24 (Stanbio Laboratory, Boerne, Tex.) and the Hgb Pro (ITC) use
reflectance photometry to measure reflected light in the test area.
Other Instruments. Other instruments that measure the The test card is composed of molded plastic with a fluid well
hematocrit include the following: that contains numerous pads impregnated with specific chemi-
ABL 77 (Radiometer, Westlake, Ohio) cal reagents. A drop of whole blood is applied to the center of
IRMA (ITC, a subsidiary of Thoratec Corporation, Edison, the well and reacts with the chemicals in the pad to produce a
N.J.) specific color that is measured from the bottom of the card.26
Gem Premier (Instrumentation Laboratory Company,
Lexington, Mass.) (Figure 14-18) Cell and Platelet Counts
Traditional cell-counting methods can be employed at the
Hemoglobin Level point of care for the analysis of WBCs, RBCs, and platelets. The
In point-of-care testing, hemoglobin level is measured by Ichor Hematology Analyzer (Helena Laboratories, Beaumont,
modified hemoglobinometers or by oximeters integrated with Tex.) performs a complete blood count along with platelet
a blood gas analyzer. The HemoCue hemoglobinometer aggregation. Another option for cell quantitation and differen-
(HemoCue, Inc., Lake Forest, Calif.) uses a small cuvette that tiation employs a buffy coat analysis method. Quantitative
contains a lysing agent and reagents to form a hemoglobin buffy coat analysis (QBC STAR, manufactured by QBC Diag-
azide, which is measured by a photometer.6 Results obtained nostics, Inc., Philipsburg, Pa.), involves centrifugation in spe-
with the instrument compare well with those produced by cialized capillary tubes designed to expand the buffy coat layer.
reference methods, but a major source of error is mixture The components (platelets, mononuclear cells, and granulo-
of blood with tissue fluid during capillary specimen collection. cytes) can be measured with the assistance of fluorescent dyes
The AVOX 1000E (ITC) measures total hemoglobin by a and a measuring device.27

SUMMARY
Although most laboratories are highly automated, the manual tests concentration of erythrocytes. The indices give an indication of the
discussed in this chapter, such as the cyanmethemoglobin method cause of an anemia.
of hemoglobin determination and centrifuge-based measurement The reticulocyte count, which is used to assess the erythropoietic
of the microhematocrit, are used as a part of many laboratories activity of the bone marrow, is accomplished through the use of
quality control and backup methods of analysis. supravital stains (e.g., new methylene blue) or by flow cytometric
The hemacytometer allows counts of any type of cell or particle methods.
(e.g., WBCs, platelets, and eosinophils) to be performed. The ESR, a measure of the settling of erythrocytes in a 1-hour
Hemoglobin determination is based on the absorbance of cyan- period, depends on the erythrocytes ability to form rouleaux.
methemoglobin at 540nm. When a spectrophotometer is used, a It is an indication of inflammation and may be used to dif
standard curve is employed to obtain the results. ferentiate various diseases or to monitor therapy for certain
The microhematocrit is a measure of packed RBC volume. disorders.
RBC indicesthe MCV, MCH, and MCHCare calculated to Point-of-care testing is often performed by nonlaboratory
determine the volume, hemoglobin content, and hemoglobin personnel.
190 PART III Routine Laboratory Evaluation of Blood Cells

Point-of-care testing is defined as diagnostic laboratory testing at For a point-of-care testing program to be successful, key elements
or near the site of patient care. such as clear administrative responsibility, well-written proce-
CLIA introduced the concept of testing site neutrality, which dures, quality control, proficiency testing, and equipment mainte-
means that it does not matter where diagnostic testing is performed nance must be present.
or who performs the test; all testing sites must follow the same Paramount to point-of-care testing is patient safety.
regulatory requirements based on the complexity of the test.
Tests are classified as waived if they are determined to be simple Now that you have completed this chapter, read again the
tests with an insignificant risk of an erroneous result. Most, but case studies at the beginning and respond to the ques-
not all, point-of-care testing is waived. tions presented.

R E V I E W Q UESTIONS
1. A 1:20 dilution of blood is made, with 3% glacial acetic 6. Calculate the MCV and MCHC for the following values:
acid as the diluent. The four corner squares on both sides RBCs5.00 1012/L
of the hemacytometer are counted for a total of 100 cells. Hb9g/dL
What is the total WBC count ( 109/L)? Hct30%
a. 0.25
MCV (fL) MCHC (g/dL)
b. 2.5
c. 5 a. 30 18
d. 10 b. 60 30
c. 65 33
2. The total WBC count is 20 109/L. Twenty-five NRBCs per d. 85 35
100WBCs are seen on the peripheral blood smear. What
is the corrected WBC count ( 109/L)? 7. What does the reticulocyte count assess?
a. 0.8 a. Inflammation
b. 8 b. Response to infection
c. 16 c. Erythropoietic activity
d. 19 d. Ability of RBCs to form rouleaux

3. If potassium cyanide and potassium ferricyanide are used 8. For a patient with the following test results, which measure
in the manual method for hemoglobin determination, the of bone marrow RBC production provides the most accu-
final product is: rate information?
a. Methemoglobin Observed reticulocyte count5.3%
b. Azide methemoglobin Hct35%
c. Cyanmethemoglobin Morphologymoderate polychromasia
d. Myoglobin a. Observed reticulocyte count
b. Corrected reticulocyte count
4. Which of the following would not interfere with the result c. RPI
when hemoglobin determination is performed by the d. ARC
cyanmethemoglobin method?
a. Increased lipids 9. Given the following values, calculate the RPI:
b. Elevated WBC count Observed reticulocyte count6%
c. Lyse-resistant RBCs Hct30%
d. Fetal hemoglobin a. 2
b. 3
5. A patient has a hemoglobin level of 8.0g/dL. According c. 4
to the rule of three, within what range would the hemato- d. 5
crit be expected?
a. 21% to 24% 10. Which of the following would be associated with an
b. 23.7% to 24.3% elevated ESR value?
c. 24% to 27% a. Osteoarthritis
d. 21% to 27% b. Polycythemia
c. Decreased globulins
d. Inflammation
CHAPTER 14 Routine and Point-of-Care Testing in Hematology: Manual and Semiautomated Methods 191

REFERENCES
1. Brecher G, Cronkite EP: Morphology and enumeration of 15. Steinvil A, Shapira I, Arbel Y, et al: Determinants of the eryth-
human blood platelets. J Appl Physiol 3:365-377, 1950. rocyte sedimentation rate in the era of microinflammation.
2. Clinical and Laboratory Standards Institute (CLSI): Reference Am J Clin Pathol 129:486-491, 2008.
and selected procedures for the quantitative determination of 16. Perkins SL: Examination of the blood and bone marrow. In
hemoglobin in blood: approved standard, 3rd ed, CLSI Doc- Greer JP, Foerster JL, Rodgers GM, et al, editors: Wintrobes
ument H15-A3, Wayne, Pa, 2000, CLSI. clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
3. Zwart A, van Assendelft OW, Bull BS, et al: Recommendations Health/Lippincott Williams & Wilkins, pp 1-20.
for reference method for haemoglobinometry in human 17. Mims MP, Prchal JT: Hematology during pregnancy. In
blood (ICSH Standard 1995) and specifications for interna- Lichtman MA, Kipps TJ, Kaushansky D, et al, editors: Williams
tional haemiglobincyanide reference preparation (4th ed). hematology, ed 7, New York, 2006, McGraw-Hill, p 101.
J Clin Pathol 49:271-274, 1996. 18. International Council for Standardization in Haematology
4. von Schenck H, Falkensson M, Lundberg B: Evaluation of Expert Panel on Blood Rheology: ICSH recommendations for
HemoCue, a new device for determining hemoglobin. Clin measurement of erythrocyte sedimentation rate. J Clin Pathol
Chem 32:526-529, 1986. 46:198-203, 1993.
5. Hemoglobin test systems. Available at: http://www.hemocue. 19. Clinical and Laboratory Standards Institute (CLSI): Reference
com/. Accessed February 20, 2010. and selected procedure for the erythrocyte sedimentation rate
6. MacLaren IA, Conn DM, Wadsworth LD: Comparison of two (ESR) test: approved standard, 4th ed, CLSI document H2-A4,
automated hemoglobin methods using Sysmex SULFOLYSER Wayne, Pa, 2000, CLSI.
and STROMATOLYSER. Sysmex J Int 1:59-61, 1991. 20. Ves-Matic line. Available at: http://www.diesse-usa.com/
7. Karsan A, MacLaren I, Conn D, et al: An evaluation of hemo- products/vesmatic/index.html. Accessed February 20, 2010.
globin determination using sodium lauryl sulfate. Am J Clin 21. Shelat S, Chacosky D, Shibutani S: Differences in erythrocyte
Pathol 100:123-126, 1993. sedimentation rates using the Westergren method and a cen-
8. Matsubara T, Mimura T: Reaction mechanism of SLS-Hb trifugation method. Am J Clin Pathol 130(1):127-130, 2008.
Method. Sysmex J (Japan) 13:206-211, 1990. 22. Centers for Medicare and Medicaid Services: Clinical labora-
9. Fujiwara C, Hamaguchi Y, Toda S, et al: The reagent tory improvement amendments (CLIA): overview. Available
SULFOLYSER for hemoglobin measurement by hematology at: http://www.cms.hhs.gov/clia/. Accessed February 22,
analyzers. Sysmex J (Japan) 13:212-219, 1990. 2010.
10. Clinical and Laboratory Standards Institute (CLSI): Proce- 23. Good laboratory practices. Available at http://www.cms.
dure for determining packed cell volume by the microhema- hhs.gov/CLIA/downloads/wgoodlab.pdf. Accessed January 8,
tocrit method: approved standard, 3rd ed, NCCLS document 2010.
H7-A3, Wayne, Pa, 2000, CLSI. 24. Abbott Point of Care Cartridge and Hematocrit/HCT and
11. Clay Adams brand SUREPREP capillary tubes, Sparks, Md, Calculated Hemoglobin/HB information sheets, in i-STAT 1
1998, Becton Dickinson Primary Care Diagnostics (product system manual. Abbott Park, Ill, revised June 11, 2008,
brochure). Abbott Laboratories.
12. Clinical and Laboratory Standards Institute (CLSI): Methods 25. Theory of operation, in Epocal system manual, rev 01, Ottawa,
for reticulocyte counting (automated blood cell counters, Ont, Canada, Epocal, Inc., pp 11-17. Available at http://
flow cytometry, and supravital dyes): approved guideline, www.epocal.com/system.html. Accessed February 20, 2010.
2nd ed, CLSI document H44-A2, Wayne, Pa, 2004, CLSI. 26. Wu J, Peterson, Mohammad A, et al: Point-of-care hemoglo-
13. College of American Pathologists: Q & A Miller disk clarifica- bin measurements by Stat-Site MHgb Reflectance Meter. Point
tion. CAP Today, April:95, 2001. Available at http://www.cap. of Care 8-11, March 2003.
org Accessed June 19, 2010. 27. QBC operators service manual, rev B, Philipsburg, Pa, 2006,
14. Koepke JF, Koepke JA: Reticulocytes. Clin Lab Haematol 8:169- QBC Diagnostics, Inc., pp 1-2.
179, 1986.

ADDITIONAL RESOURCES
College of American Pathologists, http://www.cap.org. The Joint Commission, http://www.jcaho.org.
Howerton D, Anderson N, Bosse D, et al: Good laboratory prac- Point of Care.net, http://www.pointofcare.net.
tices for waived testing sites. Available at: http://www.cdc.gov/
mmWR/PDF/rr/rr5413.pdf.
Centers for Disease Control and Prevention: Health disparities
experienced by black or African AmericansUnited States.
MMWR Morb Mortal Wkly Rep 54:1-37, 2005.
15 Examination of the Peripheral
Blood Film and Correlation with
the Complete Blood Count
Lynn B. Maedel and Kathryn Doig

OUTLINE OBJECTIVES
Peripheral Blood Films After completion of this chapter, the reader will be able to:
Specimen Collection
Peripheral Film 1. List the specimen sources and collection processes 7. Given the number of cells observed per field and the
Preparation that are acceptable for blood film preparation. magnification of the objective, apply formulas to esti-
Staining of Peripheral 2. Describe the techniques for making peripheral blood mate white blood cell (WBC) counts and platelet
Blood Films films. counts.
Peripheral Film 3. Describe the appearance of a well-prepared peri 8. Explain the effect that platelet satellitosis and clump-
Examination pheral blood film, recognize a description of a slide ing may have on automated complete blood count
Summarizing Complete that is consistent or inconsistent with that appear- (CBC) results. Recognize examples of results that
Blood Count Results ance, and troubleshoot problems with poorly pre- would be consistent with these effects.
Organization of Complete pared films. 9. Follow the appropriate course of action to recognize
Blood Count Results
4. Explain the principle, purpose, and basic method of and correct ethylenediaminetetraacetic acidinduced
Assessing Hematology
Wright staining of blood films. pseudothrombocytopenia and pseudoleukocytosis.
Results Relative to
Reference Ranges 5. Identify and troubleshoot problems that cause poorly 10. Implement a systematic approach to interpretation of
Summarizing White Blood stained blood films. CBC data that results in a verbal summary of the
Cell Parameters 6. Describe the proper examination of a peripheral numerical data and communicates the blood picture
Summarizing Red Blood blood film, including selection of the correct area, succinctly.
Cell Parameters sequence of examination, and observations to be 11. Calculate absolute WBC differential counts.
Summarizing Platelet made at each magnification. Recognize deviations
Parameters from this protocol.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A healthy looking 56-year-old man had an automated CBC RDW14.2%


performed as part of a preoperative evaluation. Results are Platelets34 109/L
shown here. Refer to reference ranges provided on the inside MPV6.6fL
front cover of this book. The peripheral blood film was examined, and the only
WBCs15.8 109/L abnormal finding was platelets in clumps.
RBCs4.93 1012/L 1. Describe the blood picture succinctly using proper
Hb14.8g/dL terminology for red blood cells, white blood cells, and
Hct45.1% platelets.
MCV91.5fL 2. What automated results should be questioned?
MCH30pg 3. What is the best course of action to handle this problem?
MCHC32.8g/dL

192
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 193

A
well-made, well-stained, and carefully examined peri
pheral blood film can provide valuable information
regarding a patients health. More can be learned from
this test than from many other routinely performed hemato-
logic tests. White blood cell (WBC) and platelet estimates can
be achieved, relative proportions of the different types of WBCs
can be obtained, and the morphology of all three cell lines can
be evaluated for abnormalities. Although routine work is now
handled by the sophisticated automated instruments found in
most hematology laboratories, skilled and talented laboratory
professionals are still essential to the reporting of reliable test
results. Proper peripheral film evaluation is quite likely to be
needed for some time.
The peripheral film evaluation is the capstone of a panel of
tests called the complete blood count (CBC) or hemogram. The Figure 15-1 Photomicrograph of platelet satellitosis.
CBC includes enumeration of cellular elements, quantitation
of hemoglobin, and statistical analyses that provide a snapshot
of cell appearances. These results can be derived using the who believe that anticoagulant-free blood is still the specimen
manual methods and calculations described in Chapter 14 or of choice for evaluation of blood cell morphology.2 Although
using the automated instruments described in Chapter 39. some artifacts can be avoided in this way, samples made from
Regardless of method, the numerical values should be consis- unanticoagulated blood pose other problems (see later).
tent with the assessment derived by examining the cells micro- Under certain conditions, use of a different anticoagulant
scopically. Careful examination of the data in a systematic way or no anticoagulant may be helpful. Some patients blood
ensures that all relevant results are noted and taken into con- undergoes an in vitro phenomenon called platelet satellitosis3
sideration in the diagnosis. when anticoagulated with EDTA. The platelets surround or
This chapter begins with a discussion of the preparation and adhere to neutrophils, which potentially causes pseudothrom-
assessment of the blood film, followed by a systematic approach bocytopenia when counting is done by automated methods
to review of the CBC, including blood film evaluation. Such (Figure 15-1).3,4 In addition, spuriously low platelet counts and
an evaluation can be applied in the hematology chapters that falsely increased WBC counts (pseudoleukocytosis) can result
follow. from EDTA-induced platelet clumping.5 Pseudoleukocytosis
occurs when platelet agglutinates are similar in size to WBCs
and automated analyzers cannot distinguish the two. The
PERIPHERAL BLOOD FILMS
platelet clumps are counted as WBCs instead of platelets.
Specimen Collection Platelet-specific autoantibodies that react best at room tem-
Sources of Specimens perature are one of the mechanisms known to cause this phe-
Essentially all specimens received for routine testing in the nomenon.6 In these circumstances, the examination of a blood
hematology section of the laboratory have been collected in film becomes an important quality control strategy, identifying
lavender (purple)topped tubes. These tubes contain diso- these phenomena so that they can be corrected before the
dium or tripotassium ethylenediaminetetraacetic acid (EDTA), results are reported to the patients chart.
which anticoagulates the blood by chelating the calcium that Problems such as these can be eliminated by re-collecting
is essential for coagulation. Liquid tripotassium EDTA is often specimens in sodium citrate tubes (light blue top) and ensur-
preferred to the powdered form because it mixes more easily ing that the proper ratio of nine parts blood to one part anti-
with blood. High-quality blood films can be made from the coagulant is observed (a properly filled tube). These new
blood in the EDTA tube, provided that they are made within specimens can be analyzed in the usual way by automated
2 to 3 hours of drawing the specimen.1 Blood films from EDTA instruments. Platelet counts and WBC counts from sodium
tubes that remain at room temperature for more than 5 hours citrate specimens must be corrected for the dilution of blood
often have unacceptable blood cell artifacts (echinocytic red with the anticoagulant, however. In a full-draw tube, the blood
blood cells [RBCs], spherocytes, necrobiotic leukocytes, and is nine tenths of the total tube volume (2.7mL of blood and
vacuolated neutrophils). Vacuolization of monocytes normally 0.3mL of sodium citrate). The dilution factor is the recipro-
occurs almost immediately with EDTA but causes no evalua- cal of the dilution (i.e., 10/9 or 1.1). The WBC and platelet
tion problems. counts are multiplied by 1.1 to obtain accurate counts. All
The main advantages of making films from blood in the other CBC parameters should be reported for the original
EDTA tube are that multiple slides can be made if necessary and EDTA tube specimen and slide.
they do not have to be prepared immediately after the blood is Another source of blood for films is from finger and heel
drawn. In addition, EDTA generally prevents platelets from punctures. In general, the films are made immediately at the
clumping on the glass slide, which makes the platelet estimate patients side. There are, however, a few limitations to this
more accurate during film evaluation. There are purists, however, procedure. First, some platelet clumping must be expected if
194 PART III Routine Laboratory Evaluation of Blood Cells

films are made directly from a drop of finger-stick or heel-stick The procedure just described is for a push-type wedge prepa-
blood or if blood is collected in heparinized microhematocrit ration. It is called push because the spreader slide is pulled into
tubes. Generally, this clumping is not enough to interfere the drop of blood and the film is made by pushing the blood
with platelet estimates if the films are made promptly before along the slide. The same procedure can be modified to produce
clotting begins in earnest. Second, only a few films can be made a pulled film. In this procedure, the spreader slide is pushed
directly from blood from a skin puncture before the site stops into the drop of blood and pulled along the length of the slide
bleeding. If slides are made quickly and correctly, however, to make the film. Although this method is less commonly used,
cell distribution and morphology should be adequate. These it also provides a satisfactory wedge preparation and is easier
problems with finger and heel sticks can be eliminated with for some individuals to perform. Other variations on the wedge
the use of EDTA microcollection tubes, such as Microtainers technique include using the 3-inch side of the slide as the
(Becton, Dickinson and Company, Franklin Lakes, N.J.) (see spreader slide or balancing the spreader slide on the fingers to
Chapter 3). avoid placing too much pressure on it. Learning to make con-
sistently good blood films takes a lot of practice but can be
Peripheral Film Preparation accomplished if one is patient and persistent.
Types of Films Features of a well-made wedge peripheral blood film
Manual Wedge Technique. The wedge film technique is 1. The film is two thirds to three fourths the length of the slide
probably the easiest to master. It is the most convenient and (Figure 15-3).
commonly used method for making peripheral blood films. 2. The film is finger shaped, very slightly rounded at the feather
This technique requires at least two 3-inch 1-inch (75-mm edge, not bullet shaped; this provides the widest area for
25-mm) clean glass slides. High-quality, beveled-edge micro- examination.
scopic slides with chamfered (beveled) corners for good lateral 3. The lateral edges of the film are visible.
borders are recommended. A few more slides may be kept 4. The film is smooth without irregularities, holes, or
handy in case a good-quality film is not made immediately. streaks.
One slide serves as the film slide, and the other is the pusher 5. When the slide is held up to the light, the thin portion
or spreader slide. They can then be reversed. It is also possible (feather edge) of the film has a rainbow appearance.
to make good wedge films by using a hemacytometer coverslip 6. The whole drop of blood is picked up and spread.
attached to a handle (pinch clip or tongue depressor) as the Figure 15-4 shows unacceptable films.
spreader.
A drop of blood (about 2 to 3mm in diameter) from a Coverslip Technique. The coverslip method of film prep-
finger, heel, or microhematocrit tube (nonheparinized for aration is an older technique that is now used only rarely for
EDTA-anticoagulated blood or heparinized for capillary blood) peripheral blood films, but it is sometimes still used for making
is placed at one end of the slide. The drop also may be deliv- bone marrow aspirate smears. The only advantage of this
ered using a Diff-Safe dispenser (Alpha Scientific Corporation, method is that it produces an excellent leukocyte distribution,
Malvern, Penn.). The Diff-Safe dispenser is inserted through which lends itself to more accurate determination of differen-
the rubber stopper of the EDTA tube, which eliminates the tial counts. For routine morphologic evaluation, however, this
need to remove the stopper.7 The size of the drop of blood is technique is neither convenient nor practical. Impeccably clean
important: too large a drop creates a long or thick film, and glass coverslips must be used.2 Labeling, transport, staining,
too small a drop often makes a short or thin blood film. The and storage of these small, breakable smears present many
pusher slide, held securely in the dominant hand at about a problems.
30- to 45-degree angle (Figure 15-2, A), is drawn back into the This technique requires that a small drop of blood or bone
drop of blood, and the blood is allowed to spread across the marrow be placed on one clean coverslip (22 22mm) and
width of the slide (Figure 15-2, B). It is then quickly and another coverslip be placed on top, with the blood allowed to
smoothly pushed forward to the end of the slide to create a spread across the two coverslips. One is then pulled across the
wedge film (Figure 15-2, C). It is important that the whole other to create two thin films, one film on each coverslip
drop be picked up and spread. Moving the pusher slide (Figure 15-5). The two films can be stained and mounted on
forward too slowly accentuates poor leukocyte distribution by a 1-inch 3-inch glass slide. When bone marrow smears are
pushing larger cells, such as monocytes and granulocytes, to made using this technique, very slight pressure is applied to
the very end and sides of the film. Maintaining an even, gentle the coverslips between the index finger and thumb to help
pressure on the slide is essential. It is also crucial to keep the spread the bone marrow spicule before the two smears are
same angle all the way to the end of the film. When the hema- pulled apart (this is sometimes called a crush preparation).
tocrit is higher than normal, as is found in patients with poly- Refining the skills required to make high-quality crush prepara-
cythemia or in newborns, the angle should be lowered (i.e., tion bone marrow smears takes practice. Too much pressure
25 degrees), so the film is not too short and thick. For extremely on the coverslips causes cell rupture, which makes morpho-
low hematocrit, the angle may need to be raised. If two or logic evaluation impossible. Inadequate pressure prevents the
three films are made, the best one is chosen for staining, and spicule from spreading to a satisfactory monolayer. Bone
the others are disposed of properly. Some laboratories make marrow crush preparations similar to this can be made using
two good films and save one unstained in case another slide regular glass slides instead of coverslips, and the smears are of
is required. equal quality.
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 195

30 45

C
Figure 15-2 Wedge technique of making a peripheral blood film.

read. The computer already has determined from the auto-


mated results whether a slide is required. Criteria for a manual
slide review are determined by each laboratory based on its
patient population. Dependent on the hematocrit reading, the
system adjusts the size of the drop of blood used and the angle
and speed of the spreader slide in making a wedge preparation.
After each blood film is prepared, the spreader slide is automati-
cally cleaned and is ready for the next blood film to be made.
Figure 15-3 Well-made peripheral blood film. Films can be produced approximately every 30 seconds. Infor-
mation such as name, number, and date for the specimen is
Automated Slide Making and Staining. The Sysmex printed on the slide. The slide is dried, loaded into a cassette,
SP-1000i (Sysmex Corporation of America, Mundelein, Ill.) is and moved to the staining position, where stain and then buffer
an automated slide making and staining system. After the and rinse are added at designated times. When staining is com-
instrument has performed a CBC for a specimen, a conveyor plete, the slide is moved to a dry position, then to a collection
moves the racked tube to the SP-1000i, where the barcode is area where it can be picked up for microscopic evaluation.
196 PART III Routine Laboratory Evaluation of Blood Cells

A B C D

E F G H
Figure 15-4 Unacceptable peripheral blood films. Slide appearances associated with the most common errors are shown, but note that a combination of
causes may be responsible for unacceptable films. A, Chipped or rough edge on spreader slide. B, Hesitation in forward motion of spreader slide. C, Spreader
slide pushed too quickly. D, Drop of blood too small. E, Drop of blood not allowed to spread across the width of the slide. F, Dirt or grease on the slide; may
also be due to elevated lipids in the blood specimen. G, Uneven pressure on the spreader slide. H, Time delay; drop of blood began to dry.

A B C

D
Figure 15-5 Coverslip method of making a peripheral blood film.
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 197

Figure 15-7 Manual Wright staining of slides. Note metallic sheen of stain
Figure 15-6 COULTER LH slide maker and stainer. (Courtesy Beckman indicating proper mixing.
Coulter, Inc., Brea, Calif.)
the thick part of the blood film may come off of the slide in
Smears made off-line, such as bone marrow smears and cyto- the staining process.
spin preparations, may be stained using this system as well. Water or drying artifact has been a long-lived nuisance to
Other blood analyzer manufacturers, such as Beckman Coulter hematology laboratories. It has several appearances. It can give
(Brea, Calif.), also have automated slide making and staining a moth-eaten look to the RBCs or it may appear as a heavily
instruments (Figure 15-6) (see Chapter 39). demarcated central pallor. It also may appear as refractive
(shiny) blotches on the RBCs. Other times, it manifests simply
Drying of Films as echinocytes (crenation) seen in the areas of the slide that
Regardless of film preparation method, before staining, all dried most slowly.
blood films and bone marrow smears should be dried as Multiple factors contribute to this problem. Humidity in the
quickly as possible to avoid drying artifact. In some laborato- air as the slide dries may add to the punched-out, moth-eaten,
ries, a small fan is used to facilitate drying. Blowing breath on or echinocytic appearance of the RBCs. It is difficult to avoid
a slide is counterproductive, because the moisture in breath drying artifact on films from extremely anemic patients because
causes RBCs to become echinocytic (crenated) or to develop of the very high ratio of plasma to RBCs. Water absorbed from
water artifact (also called drying artifact) (see later). the air into the alcohol-based stain also can contribute. Drying
the slide as quickly as possible helps, and keeping a stopper
Staining of Peripheral Blood Films tightly on the stain bottle keeps moisture out. In some labora-
Pure Wright stain or a Wright-Giemsa stain (Romanowsky tories, slides are fixed in pure, anhydrous methanol before
stain)8 is used for staining peripheral blood films and bone staining to help reduce water artifact. More recently, stain
marrow smears. These are considered polychrome stains manufacturers have used 10% volume-to-volume methanol to
because they contain both eosin and methylene blue. Giemsa minimize water or drying artifact.
stains also contain methylene blue azure. The purpose of stain-
ing blood films is simply to make the cells more visible and to Wright Staining Methods
allow their morphology to be evaluated. Manual Technique. Traditionally, Wright staining has
Methanol in the stain fixes the cells to the slide. Actual been performed over a sink or pan with a staining rack. Slides
staining of cells or cellular components does not occur until are placed on the rack, film side facing upward. The Wright
the buffer is added. The oxidized methylene blue and eosin stain may be filtered before use or poured directly from the
form a thiazine-eosinate complex, which stains neutral bottle through a filter onto the slide. It is important to flood
components. The buffer that is added to the stain should be the slide completely. The stain should remain on the slide at
0.05M sodium phosphate (pH 6.4) or aged distilled water least 1 to 3 minutes to fix the cells to the glass. Then an approxi-
(distilled water placed in a glass bottle for at least 24 hours; mately equal amount of buffer is added to the slide. Surface
pH 6.4 to 6.8). Staining reactions are pH dependent. Free tension allows very little of the buffer to run off. The laboratory
methylene blue is basic and stains acidic (and basophilic) cel- professional can blow gently on the slide to mix the aqueous
lular components, such as ribonucleic acid (RNA). Free eosin buffer and the alcohol stain. A metallic sheen (or green scum)
is acidic and stains basic (and eosinophilic) components, such should appear on the slide if mixing is correct (Figure 15-7).
as hemoglobin and eosinophilic granules. Neutrophils are so More buffer can be added if necessary. The mixture is allowed
named because they have cytoplasmic granules that have a to remain on the slide for 3 minutes or more (bone marrow
neutral pH and pick up some staining characteristics from both smears take longer to stain than peripheral blood films).
stains. The slides must be completely dry before staining, or The timing may be adjusted to produce the best staining
198 PART III Routine Laboratory Evaluation of Blood Cells

characteristics. When staining is complete, the slide is rinsed Automated Slide Stainers. Numerous automated slide
with a steady but gentle stream of neutral-pH water, the back stainers are commercially available. For high-volume laborato-
of the slide is wiped to remove any stain residue, and the slide ries, these instruments are essential. Once they are set up and
is air-dried in a vertical position. Coverslip-type blood films loaded with slides, staining proceeds without operator atten-
must be stained by the manual method. These films are placed tion. In general, it takes about 5 to 10 minutes to stain a batch
on evacuated test tube rubber stoppers on the staining rack of slides. The processes of fixing/staining and buffering are
over a sink (Figure 15-8). similar in practice to those of the manual method. The slides
Use of the manual Wright staining technique is desirable may be automatically dipped in stain and then in buffer and
for staining peripheral blood films containing very high WBC a series of rinses (Midas II, EM Science, Gibbstown, N.J. [Figure
counts, such as the films from leukemic patients. As with bone 15-9]) or propelled along a platen surface by two conveyor
marrow smears, the time can be lengthened easily to enhance spirals (Hema-Tek, Siemens Healthcare Diagnostics, Inc., Deer-
the staining required by the increased numbers of cells. Under- field, Ill. [Figure 15-10]). In the Hema-Tek device, stain, buffer,
staining is common when a leukemia slide is placed on an and rinse are pumped through holes in the platen surface,
automated slide stainer. The main disadvantages of the manual flooding the slide at the appropriate time. Film quality and
technique are the increased risk of spilling the stain and the color consistency are usually good with any of these instru-
longer time required to complete the procedure. This tech- ments. Some commercially prepared stain, buffer, and rinse
nique is best suited for low-volume laboratories. packages do vary from lot to lot or manufacturer to manufac-
turer, so testing is recommended. Some disadvantages of the
dip-type batch stainers are that (1) stat slides cannot be added
to the batch once the staining process has begun, and (2)
working or aqueous solutions of stain are stable for only 3 to
6 hours and need to be made often. Stat slides can be added
at any time to the Hema-Tek stainer, and stain packages are
stable for about 6 months.

Quick Stains. Quick stains, as the name implies, are fast


and easy. The whole process takes about 1 minute. The stain
is purchased in a bottle as a modified Wright or Wright-Giemsa
stain. The required quantity can be filtered into a Coplin jar or
a staining dish, depending on the quantity of slides to be
stained. Aged distilled water is used as the buffer. Stained slides
are given a final rinse under a gentle stream of tap water and
allowed to air-dry. It is helpful to wipe off the back of the slide
with alcohol to remove any excess stain. Quick stains are con-
venient and cost effective for low-volume laboratories, such as
Figure 15-8 Manual Wright staining of coverslips. clinics and physician office laboratories, or whenever rapid

Figure 15-9 Midas II slide stainer. (Courtesy EM Science, Gibbstown, N.J.).


CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 199

BOX 15-1 Troubleshooting Poorly Stained Blood Films

First Scenario
Problems
RBCs appear gray.
WBCs are too dark.
Eosinophil granules are gray, not orange.

Causes
Stain or buffer too alkaline (most common)
Inadequate rinsing
Prolonged staining
Heparinized blood sample

Figure 15-10 Hema-Tek 2000 slide stainer. ( 2009 Siemens Health- Second Scenario
care Diagnostics, Inc. Photo courtesy Siemens Healthcare Diagnostics.) Problems
RBCs are too pale or are red.
WBCs are barely visible.
turnaround time is essential. Quality is often a concern with
quick stains. With a little time and patience in adjusting the Causes
staining and buffering times, however, color quality can be Stain or buffer too acidic (most common)
acceptable. Underbuffering (too short)
Over-rinsing
Features of a Well-Stained Peripheral Blood Film. Pro RBC, Red blood cell; WBC, white blood cell.
perly staining a peripheral blood film is just as important as
making a good film. Macroscopically, a well-stained blood film
should be pink to purple. Microscopically, the RBCs should may be seen on the film. A grainy appearance to the film may
appear orange to salmon pink, and WBC nuclei should be indicate RBC agglutination, as found in cold hemagglutinin
purple to blue. The cytoplasm of neutrophils should be pink diseases. In addition, holes all over the film could mean that
to tan with violet or lilac granules. Eosinophils should have the patient has increased lipid levels, and some of the auto-
bright orange refractile granules. Faulty staining can be trouble- mated CBC parameters should be rechecked for interferences
some for reading the films, causing problems ranging from from lipemia. Markedly increased WBC counts and platelet
minor shifts in color to the inability to identify cells and assess counts can be detected from the blue specks out at the feather
morphology. Trying to interpret a poorly prepared or poorly edge. Valuable information might be obtained before the
stained blood film is extremely frustrating. If possible, a newly evaluator looks through the microscope.
stained film should be studied. Hints for troubleshooting
poorly stained blood films are provided in Box 15-1. Microscopic Examination
The best staining results are obtained on fresh slides, The microscope should be adjusted correctly for blood film
because the blood itself acts as a buffer in the staining process. evaluation. The light from the illuminator should be properly
Slides stained after 1 week or longer turn out too blue. In addi- centered, the condenser should be almost all the way up and
tion, specimens that have increased levels of proteins (i.e., adjusted correctly for the magnification used, and the iris dia-
globulins) produce bluer-staining blood films, even when phragm should be opened to allow a comfortable amount of
freshly stained. light to the eye. Many individuals prefer to use a neutral density
filter over the illuminator to create a whiter light from a tung-
Peripheral Film Examination sten light source. If the microscope has been adjusted for
Microscopic blood film review is essential whenever instrument Koehler illumination, all these conditions should have been
analysis indicates that specimen abnormalities exist. The labo- met (see Chapter 4).
ratory professional evaluates the platelet and WBC count and
differential, along with WBC, RBC, and platelet morphology. 10 Objective Examination. Blood film evaluation begins
using the 10 or low-power objective lens (total magnification
Macroscopic Examination = 100). Not much time needs to be spent at this magnifica-
Examining the film before placing it on the microscope stage tion. However, it is a common error to omit this step altogether
sometimes can give the evaluator an indication of abnormali- and go directly to the higher-power oil immersion lens. At the
ties or test results that need rechecking. A film that is bluer low-power magnification, overall film quality, color, and dis-
overall than normal may indicate that the patient has increased tribution of cells can be assessed. The feather edge and lateral
blood proteins, as in multiple myeloma, and that rouleaux edges can be checked quickly for WBC distribution. The
200 PART III Routine Laboratory Evaluation of Blood Cells

presence of more than four times the number of cells per field 100 oil immersion objective also can be accomplished by
at the edges or feather compared with the monolayer area of experienced morphologists using a 50 oil immersion objec-
the film indicates that the film is unacceptable (i.e., a snow- tive. The larger field of view allows more cells to be evaluated
plow effect), and the film should be remade. Under the 10 faster. Examination at this power is especially efficient for vali-
objective, it is possible to check for the presence of fibrin dating or verifying instrument values when a total microscopic
strands; if they are present, the sample should be rejected, and assessment of the film is not needed. Particular cell features
another one should be collected. RBC distribution can be that may require higher magnification can be assessed by
noted as well. Rouleaux formation or RBC agglutination is easy moving the parfocal 100 objective into place, then returning
to recognize at this power. The film can be scanned quickly for to the 50 objective to continue the differential. The WBC
any large abnormal cells, such as blasts, reactive lymphocytes, estimate also can be performed with the 50 objective,
or even unexpected parasites. Finally, the area available for but the multiplication factor would be 3000. The observer
suitable examination can be assessed. should conform to the estimation protocol of the particular
laboratory.
40 Objective Examination. The next step is using the
40 high dry objective lens (total magnification = 400). At Optimal Assessment Area. The tasks described espe-
this magnification, it is easy to select the correct area of the film cially for the 100, 40, and 50 objectives need to be per-
in which to begin the differential count and to evaluate cellular formed in the best possible area of the peripheral blood film.
morphology. The WBC estimate also can be performed at this That occurs between the thick area, or heel, where the drop
power. To perform a WBC estimate, the evaluator selects an of blood was initially placed and spread, and the very thin
area in which two or three RBCs overlap, but most RBCs are feather edge. Microscopically, the RBCs are uniformly and
separated from one another. The average number of WBCs per singly distributed, with few touching or overlapping, and have
high-power field is multiplied by 2000 to get an adequate their normal biconcave appearance (central pallor) (Figure
approximation of the WBC count. If, after 8 to 10 fields are 15-11). An area that is too thin, in which there are holes in the
scanned, it is determined that there are four to five WBCs per film and the RBCs look flat, large, and distorted, is unaccept-
field, this would yield a WBC estimate of 8000 to 10,000/mm3 able. A too-thick area also distorts the RBCs by piling them on
(8 to 10 109/L). This technique can be helpful for internal top of one another like rouleaux (Figure 15-12). WBCs are
quality control, although there are inherent errors in the similarly distorted, which makes morphologic evaluation more
process. Just being too deep (toward the heel) on the slide or difficult and classification potentially incorrect. When the
too shallow (toward the feather) affects the estimates. In addi- correct area of a specimen from a patient with a normal RBC
tion, field diameters may vary among microscope manufactur- count is viewed, there are generally about 200 to 250RBCs per
ers. Checking for a discrepancy between the estimate and the 100 oil immersion field.
instrument WBC count makes it easier to discover problems Although described in Chapter 4, a common problem
such as a film made using the wrong patients blood sample encountered with the oil immersion objective lens is worth
or a mislabeled film. In many laboratories, WBC estimates are mentioning again. If the blood film was in focus under the 10
performed on a routine basis; in others, these estimates are and 40 objectives but is impossible to bring into focus under
performed only as needed to confirm instrument values. More the 100 objective, the slide is probably upside down. The
detailed instructions are given in Chapter 14. 100 objective does not have sufficient depth of field to focus
through the slide. The oil must be completely removed before
100 Oil Immersion Objective Examination. The 100 the film is put on the stage right side up.
oil immersion objective provides the highest magnification on
most standard binocular microscopes (10 eyepiece 100
objective lens = 1000 magnification). The WBC differential
count generally is performed using the 100 oil immersion
objective. Performing the differential normally includes count-
ing and classifying 100WBCs to obtain percentages of WBC
types. The RBC, WBC, and platelet morphology evaluation and
the platelet estimate also are executed under the 100 oil
immersion objective lens. At this magnification segmented
neutrophils can be readily differentiated from bands. RBC
inclusions, such as Howell-Jolly bodies, and WBC inclusions,
such as Dhle bodies, can be seen easily if present. Reactive or
abnormal cells are enumerated under the 100 lens as well. If
present, nucleated RBCs (NRBCs) are counted and reported as
NRBCs per 100WBCs (see Chapter 14).

50 Oil Immersion Objective Examination. The WBC


differential and morphology examinations described for the Figure 15-11 Photomicrograph of good area of film.
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 201

Figure 15-14 Laboratory differential tally counter.

monocytes, 3% eosinophils. The evaluator always should


check to ensure that the sum of the percentages is 100.
Performing 100-cell differentials on extremely low WBC
counts can become tedious and time consuming, even when
the 50 oil immersion objective is used. In some laboratories,
B
the WBCs are concentrated by centrifugation, and buffy coat
Figure 15-12 Photomicrograph of areas too thin (A) and too thick (B) to smears are made. This practice is helpful for examining the
read. morphology of the cells; however, it is not recommended for
performing differentials because of possible errors in cell dis-
tribution from centrifugation. In other laboratories evaluators
may perform a 50-cell differential and multiply the results by
2 to get a percentage. The accuracy of this practice is question-
able, and it should be avoided if possible. In some laboratories
the buffy coat smear is examined for the presence of blasts, but
no differential is performed. It is important to include the side
margins of the blood smear in any differential so that the larger
cells, such as monocytes, reactive lymphocytes, and immature
Figure 15-13 Battlement pattern for performing a differential count. cells, are not excluded.
In addition to counting the cells, the evaluator assesses their
appearance. If present, WBC abnormalities such as toxic granu-
Performance of the White Blood Cell Differential. Fewer lation, Dhle bodies, reactive lymphocytes, and Auer rods (see
manual differentials are performed today because of the supe- Chapter 28) are evaluated and reported. The exact method by
rior accuracy of automated differentials and because of cost which these are reported varies from laboratory to laboratory.
and time constraints. When indicated, however, the manual Reactive lymphocytes may be reported as a separate percentage
differential always should be performed in a systematic of the 100 cells, as a percentage of the total number of
manner. When the correct area has been selected, use of a back- lymphocytes, or semiquantitatively (occasional to many).
and-forth serpentine, or battlement, track pattern is preferred Toxic granulation generally is reported as present or is reported
to minimize distribution errors (Figure 15-13)9 and ensure that semiquantitatively (slight to marked, or 1+ to 3+). Stan-
each cell is counted only once. One hundred WBCs are counted dardization of this process has been difficult. Regardless of
and classified through the use of push-down button counters reporting format, each laboratory should establish criteria for
(Figure 15-14) or newer computer-interfaced touch pads quantifying microscopic cell morphology.
(Figure 15-15). To increase accuracy, it is advisable to count at Because the differential alone provides only partial informa-
least 200 cells when the WBC count is higher than 40 109/L. tion, the absolute cell counts are calculated for each cell type
The results are reported as percentages: for example, 54% (see later) in some laboratories. Automated differentials
segmented neutrophils, 6% bands, 28% lymphocytes, 9% already include this information.
202 PART III Routine Laboratory Evaluation of Blood Cells

Figure 15-15 Biovation differential counter. (ISYS/Biovation, Orlando, Fla.)

Red Blood Cell Morphology. Evaluation of RBC mor- Regardless of whether or not an official estimate is made,
phology is an important part of the blood film examination verification of the instrument platelet count should be included
and includes an assessment of cell size, variability in size in the overall examination. Blood film examination also
(anisocytosis), cell color, cell shape (poikilocytosis), and cel- includes an assessment of the morphology of the platelets,
lular inclusions (see Chapter 18). Some laboratories use specific including size as well as granularity and overall appearance.
terminology for reporting the degree of abnormal morphology, As mentioned in Chapter 4, immersion oils with different
such as slight, moderate, or marked, or use a scale from viscosities do not mix well. If slides are taken to another micro-
1+ to 3+. Other laboratories provide a summary statement scope for review, oil should be wiped off first.
regarding the overall RBC morphology that is consistent with
the RBC indices. The latter method is becoming more popular
SUMMARIZING COMPLETE BLOOD
with the increased computer interfacing in most laboratories.
COUNT RESULTS
Regardless of the reporting method used, the microscopic RBC
morphology assessment should be congruent with the informa- To this point, this chapter has focused on slide preparation and
tion given by the automated processor. If not, further investiga- performance of a differential cell count. The differential is only
tion is needed. the capstone, however, of a panel of tests collectively called the
CBC that includes many of the routine tests described in
Platelet Estimate. As previously mentioned, the platelet Chapter 14. Now that the testing for the component parts has
estimate is performed under the 100 oil immersion objective been described, interpretation of the results for the total panel
lens. In an area of the film where the RBCs barely touch, the can be discussed.
number of platelets in 10 oil immersion fields is counted. The The CBC has evolved over time to the typical test panel
average number of platelets per oil immersion field times reported today, including assessment of WBCs, RBCs, and
20,000 approximates the platelet count per mm3. For example, platelets. The CBC provides information about the hematopoi-
12 to 16 platelets per oil immersion field equals about 280,000 etic system, but because abnormalities of blood cells can be
platelets/mm3 (280 109/L) and is considered adequate. A caused by diseases of other organ systems, the CBC also plays
rougher estimate states that if there are 7 to 25 platelets per oil a role in screening of those organs for disease. The CBC pro-
immersion field, the platelet count is adequate, provided that vides such valuable information about a patients health status
there are approximately 200RBCs per oil immersion field. In that it is among the most frequently ordered laboratory tests
situations in which the patient is anemic or has erythrocytosis, performed by medical laboratory scientists and laboratory
however, the relative proportion of platelets to RBCs is altered. technicians.
In these instances, a more involved formula for platelet esti- The process of interpreting the CBC test results has two
mates may be used: phases. In phase 1, the numbers and descriptions generated by
the testing are summarized using appropriate terminology. This
Average no. of platelets field total RBC count summary provides a verbal picture of the numbers that is easy
200 RBCs field to communicate to the physician or another laboratorian. It is
much more convenient to be able to say, The patient has a
(200 is the average number of RBCs per oil immersion field in microcytic anemia than to say, The hemoglobin was low, and
the optimal assessment area) the mean cell volume was also low. Phase 2 of interpretation
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 203

is to recognize a pattern of results consistent with various dis- 3. WBC differential count values expressed as the actual
eases and to be able to narrow the diagnosis for the given number of each type of cell (e.g., neutrophils 109/L)
patient or perhaps even to pinpoint it so that appropriate absolute counts
follow-up testing can be recommended. 4. WBC morphology
The following discussion focuses on phase 1 of CBC
interpretationhow to collect the pertinent information and Step 1
summarize it. Phase 2 of the interpretation is the essence of Start by ensuring that there is an accurate WBC count. Modern
the remaining chapters of this text on various hematologic instruments are able to eliminate nucleated RBCs that falsely
conditions or other metabolic conditions that have an impact increase the WBC count. However, manual WBC results must
on the hematologic system. be corrected mathematically to eliminate the contribution of
the NRBCs (see Chapter 14).
Organization of Complete Blood
Count Results Step 2
Today, most laboratorians perform CBCs using sophisticated Look at the total WBC count. When the count is elevated, it is
automated analyzers as described in Chapter 39, but the com- called leukocytosis. When the WBC count is low, it is called
ponent tests can be performed using the manual methods leukopenia. As described later, increases and decreases of WBCs
described in Chapter 14. The blood film assessment described are associated with infections and conditions such as leuke-
in this chapter is also part of a CBC. The CBC panel is essen- mias. Because there is more than one type of WBC, increases
tially divided into WBC, RBC, and platelet parameters. and decreases in the total count are usually due to changes in
For phase 2 interpretation, it is sometimes important to one of the subtypes. Determining which one is the next step.
look at all three groupings of the CBC results; at other times,
only one or two may be of interest. If a patient has an infection, Step 3
the WBC parameters may be the only ones of interest. If the Examine the relative differential counts for a preliminary
patient has anemia, all three setsWBCs, RBCs, and platelets assessment of which cell lines are affected. The relative differ-
may require assessment. Generally, all the parameters inter- ential count is reported in percentages. The proportion of each
preted together provide the best information, so a complete cell type can be described by its relative number (i.e., percent)
summary of the results should be generated. and compared with its reference range. Then it is described
using appropriate terminology, such as a relative neutrophilia,
Assessing Hematology Results Relative to which is an increase in neutrophils, or a relative lymphopenia,
Reference Ranges which is a decrease in lymphocytes. The terms used for increases
Proper performance of the phase 1 summarization of test and decreases of each cell type are provided in Table 15-1.
results requires comparison of the patient values with the If the total WBC count is outside the range or if any of the
normal reference ranges. Examination of the table of reference relative values are outside the range, further analysis of the
values for the CBC on the inside cover of this text shows that WBC differential is needed. If the proportion of one of the cell
there are different reference ranges for men and women, par- types increases, then the proportion of others must decrease,
ticularly for the RBC parameters. There also are different ranges because the proportions are relative to one another. The second
for children of different ages, with the WBC changes the most cell type may not have changed in actual number at all,
notable. It is important to select the appropriate set of reference however. The way to assess this accurately is with absolute dif-
values in hematology for the sex and age of the patient. ferential counts.
Several strategies can help in determining the significance of
the results. First, if the results are very far from the reference Step 4
range, it is more likely that they are truly outside the range and If not reported by the instrument, absolute counts can be cal-
represent a pathologic process. Second, if two or more diagnosti- culated easily using the total WBC count and the relative dif-
cally related parameters are slightly or moderately outside the ferential. Multiply each relative cell count (i.e., percentage) by
range in the same direction (both high or both low), this suggests
that the results are clinically significant and associated with some
pathologic process. Because some healthy individuals always
TABLE 15-1 Terminology for Increases and Decreases
have results slightly outside the reference range, the best com-
in White Blood Cells
parison for their results is not the reference range, but their own
results from a prior time when they were known to be healthy. Cell Type Increases Decreases
Neutrophil Neutrophilia Neutropenia
Summarizing White Blood Cell Parameters Eosinophil Eosinophilia N/A
The WBC-related parameters of a routine CBC include the Basophil Basophilia N/A
following: Lymphocyte Lymphocytosis Lymphopenia (lymphocytopenia)
1. Total WBC count (WBCs 109/L) Monocyte Monocytosis Monocytopenia
2. WBC differential count values expressed as percentages
relative counts N/A, Not applicable because the reference range begins at or near zero.
204 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 15-2 Comparison of Relative and Absolute White Blood Cell Counts
Complete Blood Patient Interpretation Relative
Count Parameter Value Reference Range to Reference Range Summary
White blood cell count 13.6 4.3-10.8 10 /L
9
Elevated Leukocytosis
Relative differential
Neutrophils 67 48-70% WRR
Lymphocytes 26 18-42% WRR
Monocytes 3 1-10% WRR
Eosinophils 3 1-4% WRR
Basophils 1 0-2% WRR
Absolute differential
Absolute neutrophils 9.1 2.4-8.2 109/L Elevated Absolute neutrophilia
Absolute lymphocytes 3.5 1.4-4 109/L WRR
Absolute monocytes 0.4 0.1-1.2 109/L WRR

WRR, Within reference range.

the total WBC count and by so doing determine the absolute


count for each. BOX 15-2 Summarizing Neutrophilia When Young
Examine the set of WBC parameters from a CBC shown in Cells Are Present
Table 15-2. On first inspection, one may look at the WBC count A subtle convention in assessing the differential counts has to do
and recognize that a leukocytosis is present, but it is important with the presence of young cell types. The young cells of the granu-
to determine what cell line is causing the increased count. In locytic series (e.g., bands, metamyelocytes) typically are lumped
this case, the cells are all within normal reference ranges rela- together with the mature neutrophils in judging whether neutrophilia
tive to one another. There is no indication as to which cell line is present. For example, look at the following differential and the
could be causing the increase in total numbers of WBCs. reference ranges provided:
When each relative number (e.g., neutrophils at 0.67 or White blood cells10.8 109/L
67%) is multiplied by the total WBC count (13.6 109/L), the Neutrophils (also called granulocytes)65 (48-70%)
absolute numbers indicate that the neutrophils are elevated Bands18 (0-10%)
(9.1 109/L compared with the reference range provided). The Lymphocytes13 (18-42%)
absolute lymphocyte count (3.5 109/L) is still normal. Given Monocytes3 (1-10%)
this information, these results can be described as showing a Eosinophils1 (1-4%)
leukocytosis with only an absolute neutrophilia, and the Basophils0 (0-2%)
overall increase in the WBC count is due to an increase only Although the mature neutrophils are within the reference range,
in neutrophils. This description provides a concise summary the bands exceed their range. The total of the two (65% + 18%)
of the WBC counts without the need to refer to every type of exceeds the upper limit of neutrophilic cells even when the two ranges
cell. Box 15-2 extends this concept to a convention used to are combined (70% + 10%), so these results would be described
describe the neutrophilic cells. as a neutrophilia (or granulocytosis) even though the neutrophil value
When the absolute numbers of each of the individual cell itself is within range. See steps 4 and 5 under Summarizing White
types are totaled, the sum equals the WBC count (slight differ- Blood Cell Parameters for how to communicate the increase in bands.
ences may occur due to rounding, as in the example). This is
a method for checking whether the absolute calculations are
correct. Absolute counts may be obtained directly from auto- may indicate infections or malignancies such as leukemia. For
mated analyzers, which count actual numbers (i.e., produce neutrophilic cells, there is a unique term that refers to the pres-
absolute counts) and calculate relative values. Some laborato- ence of too many bands or cells younger than bands: left shift
ries do not report the absolute counts, so being able to calcu- (Box 15-3).
late them is important. When young lymphocytic or monocytic cells are present,
As will be evident in later chapters, the findings in this they can be reported in the differential as prolymphocytes,
particular example point toward a bacterial infection. Had lymphoblasts, promonocytes, or monoblasts. Young eosino-
there been an absolute lymphocytosis, a viral infection would phils and basophils are typically just called immature and are
be likely. not staged.

Step 5 Step 6
Each cell line should be examined for immature cells. Young Any abnormalities of appearance are reported in the morphol-
WBCs are not normally seen in the peripheral blood, and they ogy section of the report. For WBCs, abnormal morphologic
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 205

TABLE 15-3 Interpretation of Mean Cell Volume Values


BOX 15-3 Origin of the Phrase Left Shift Using the Wintrobe Terminology
The origin of the phrase left shift is a 1920s publication by Josef
Arneth in which neutrophil maturity was correlated with segment Mean Cell Volume Value Wintrobe Description
count. A graphical representation was made, and the fewer the Within reference range Normocytic
segments, the further left was the median, hence left shift. This was Lower than reference range Microcytic
called the Arneth count or Arneth-Schilling count and was aban- Higher than reference range Macrocytic
doned around the time Arneth died in 1955, but the term left shift
lived on as an indicator of infection.
The MCV is expected to be within the established reference
range (approximately 80 to 100fL), and the RBC morphology
features that would be noted include changes in overall cellular is expected to be normal (normocytic). For a patient with
appearance, such as reactive lymphocytes; cytoplasmic inclu- anemia, classifying the anemia morphologically by the MCV
sions such as Auer bodies; and nuclear abnormalities like narrows the range of possible causes to microcytic, normocytic,
hypersegmentation. The clinical significance of these types of or macrocytic anemias.
changes is discussed in Chapter 28.
To summarize the WBC parameters, begin with an accurate Step 3
total WBC count, followed by the relative differential, or prefer- Examine the MCHC to evaluate how well the cells are
ably the absolute counts, noting whether any abnormal young filled with hemoglobin. If the MCHC is below the reference
cells are present in the blood. Finally, note the presence of any range, the cells are called hypochromic, which meaning too
abnormal morphology or inclusions. little color. This correlates with a larger central pallor (hypo-
chromia) when the cells are examined on a Wright-stained
Summarizing Red Blood Cell Parameters film. Otherwise, the cells are said to be normochromic
RBC parameters are as follows: and should show the typical pallor in the central third of
1. RBC count (RBCs 1012/L) the cells.
2. Hb (g/dL) It is possible for the MCHC to be elevated in two situations.
3. Hct (% or L/L) A slight elevation may be seen when cells are spherocytic
4. Mean cell volume (MCV, fL) (MCHC over 37). They retain roughly normal volume but have
5. Mean cell Hb (MCH, pg) decreased surface area and lose some water. So the hemoglobin
6. Mean cell Hb concentration (MCHC, % or g/dL) is slightly more concentrated than usual. A more dramatic
7. RBC distribution width (RDW, %) increase in MCHC (values even as high as 60) can be caused
8. Morphology by analytical problems, often associated with patient sample
problems that falsely elevate hemoglobin measurement.
Step 1 Common problems of this type include interference from
Examine the hemoglobin (or hematocrit) for anemia or lipemia, icterus, or grossly elevated WBC counts. Each of these
polycythemia. Anemia is the more common condition. If interferes with the spectrophotometric measurement of hemo-
the RBC morphology is relatively normal, three times the globin, thus falsely elevating the hemoglobin and affecting the
hemoglobin approximates the hematocrit; this is the rule of calculation of the MCHC (see Chapter 14).
three (see Chapter 14). If the rule of three holds, the expecta-
tion is that the following assessments will find normal RBC Step 4
parameters. If the rule of three fails and all test results are The RDW is determined from the histogram of RBC volumes.
reliable, further assessment should uncover some patient RBC Briefly, when the volume of the RBCs is variable (more small
abnormalities. cells, more large cells, or both), the histogram becomes wider.
Hemoglobin concentration is a more reliable indicator of The width of the histogram, the RDW, is reflected statistically
anemia than is the hematocrit, because the hematocrit can be as the coefficient of variation (CV) or the standard deviation
influenced by the size of the RBCs. Hemoglobin concentration (SD). Several analyzer manufacturers provide a CV and an SD,
is a more direct indicator of the ability of the blood to carry and the operator can select which to report.
oxygen. The RDW provides information about the presence and
degree of anisocytosis. What is important is increased values
Step 2 only, not decreased values. If an RDW-CV reference range is
When the hemoglobin and hematocrit values have been 11.5% to 14.5% and a patient has an RDW-CV of 20.6%, the
inspected and the rule of three applied, the next RBC parameter patient has a more heterogeneous RBC population with more
that should be evaluated is the MCV (see Chapter 14). This variation in cell volume (anisocytosis). If the RDW is elevated,
value provides the average RBC volume. The MCV can be cor- a notation about anisocytosis is expected in the morphologic
related with the morphologic appearance of the cells using evaluation of the blood film. Many individuals find that using
the classification first introduced by Wintrobe a century ago the MCV along with the RDW provides the most helpful infor-
(Table 15-3). mation (see Chapters 18 and 39).
206 PART III Routine Laboratory Evaluation of Blood Cells

Step 5 this reason it is less often used than the MCV and MCHC. In
Examine the morphology for pertinent abnormalities. When- the instances in which the MCH does not follow the MCV, the
ever anemia is indicated by the RBC parameters reported by MCHC detects the discrepancy between size and hemoglobin
the analyzer and potential abnormal RBC morphology is content of the cell. The MCH is not crucial to the assessment
suggested by the indices and rule of three, a Wright-stained of anemia when the other parameters are provided. In summary,
peripheral blood film must be examined. Abnormalities when evaluating the RBC parameters of the CBC, examine the
include (1) abnormal size, (2) abnormal shape, (3) inclusions, hemoglobin first, then look at the MCV, RDW, and MCHC.
(4) young RBCs, (5) abnormal color, and (6) abnormal Finally, take note of any abnormal morphology.
arrangement (see Chapter 18). The blood film also serves as
quality control, because the morphologic characteristics seen Summarizing Platelet Parameters
through the microscope (e.g., microcytosis, anisocytosis) The platelet parameters of the CBC are as follows:
should be congruent with the results provided by the analyzer. 1. Platelet count (platelets 109/L)
When they do not agree, further investigation is necessary. 2. Mean platelet volume (MPV, fL)
If everything in the morphology is normal, by convention, 3. Morphology
no notation regarding morphology is included. Therefore, any
notation in the morphology section requires scrutiny. All RBC- Step 1
related abnormal morphologic findings should be noted, The platelet count should be examined for increases (throm-
including specific poikilocytosis (abnormal shape) and the bocytosis) or decreases (thrombocytopenia) outside the estab-
presence of RBC inclusions, such as Howell-Jolly bodies. One lished reference range. A patient who has unexplained bruising
of the major challenges is in determining when the amount or or bleeding may have a decreased platelet count. The platelet
degree of an abnormality is worth mentioning at all (i.e., when count should be assessed along with the WBC count and
it should be reported). Laboratories strive for some standard- hemoglobin concentration to determine whether all three
ization of reporting. All laboratories must have good criteria are decreased (pancytopenia) or increased (pancytosis). Pan-
and staff whose evaluations are consistent with one another cytopenia is clinically significant because it can indicate a
and with the standardized criteria used in that facility. Gener- possible developing acute leukemia or aplastic anemia.
ally a semiquatitative method is used that employs terms such Pancytosis frequently is associated with a diagnosis of poly
as slight, moderate, and marked or the numbers 1+, 2+, cythemia vera.
and 3+. Numerical ranges representing the reporting unit are
defined; for example, three to six spherocytes per oil immer- Step 2
sion field might be reported as 2+ spherocytes. Compare the instrument-generated MPV with the MPV refer-
The presence of young RBCs suggests that the bone marrow ence range, 6.9 to 10.2fL, and with the platelet diameter
is attempting to respond to an anemia. Polychromasia on the observed on the peripheral blood film. An elevated MPV
peripheral blood film indicates bone marrow response. This is should correspond with increased platelet diameter, just as an
manifested by the bluer color of reticulocytes that have entered elevated MCV reflects macrocytosis. In platelet consumption
the bloodstream earlier than usual in the bodys attempt to disorders such as immune thrombocytopenic purpura, an ele-
improve oxygen-carrying capacity. If the anemia is quite severe, vated MPV, accompanied by platelets 6m or larger in diam-
NRBCs also may be present. As noted earlier, it is also impor- eter, reflects bone marrow release of early stress or reticulated
tant to recognize NRBCs, because they may falsely elevate the platelets, evidence for bone marrow compensation (see Chapter
WBC count. 13).10,11 Comparing platelet diameter by visual inspection with
A better way to assess replacement erythropoiesis is with the MPV is a recommended quality control step; however, the MPV
reticulocyte count and subsequent calculation of the reticulo- has a wide percent CV, which reflects interindividual variation
cyte production index, if appropriate. The reticulocyte count is and platelet swelling in EDTA and reduces its clinical effective-
not normally part of the CBC, although it is now performed ness.12 Some instruments identify and record a reticulated
on the same analyzers. If the reticulocyte count is available platelet count using nucleic acid dye, analogous to a reticulo-
with the CBC, its interpretation can improve the assessment of cyte count.
young RBCs (see Chapter 14).
Step 3
Step 6 Examine platelet morphology and platelet arrangement.
Examine the remaining RBC count and MCH. On a practical Although the MPV can recognize abnormally large platelets,
level, the RBC count is not the parameter used to judge anemia, the morphologic evaluation also notes this. Some laboratories
because there are some types of anemia, such as the thalasse- distinguish between large platelets (two times normal size) and
mias, in which the RBC count is normal or even elevated. Thus giant platelets (more than twice as large as normal) or compare
the assessment of anemia would be missed by relying only on platelet size to RBC size.
the RBC count. However, this inconsistency (low hemoglobin Additional morphologic descriptors include terms for
concentration and high RBC count) is helpful diagnostically. reporting granularity, which is most important if missing, and
The MCH follows the MCV; that is, smaller cells necessarily in this case the platelets are described as hypogranular or
hold less hemoglobin, whereas larger cells can hold more. For agranular. Sometimes the abnormalities are too variable to
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 207

BOX 15-4 Applying a Systematic Approach to Complete Blood Count Summarization


A specimen from an adult male patient yields the following CBC results (refer to the reference ranges inside the front cover of this book):
WBCs3.20 109/L
RBCs2.10 1012/L
Hb8.5g/dL
Hct26.3%
MCV125fL
MCH40.5pg
MCHC32.3g/dL
RDW20.6%
Platelets115 109/L
Differential in percentages:
Neutrophils43
Bands2
Lymphocytes45
Monocytes10
Morphology: hypersegmentation of neutrophils, anisocytosis, macrocytes, oval macrocytes, occasional teardrop, Howell-Jolly bodies, and basophilic
stippling
The step-by-step assessment of the WBCs indicates that the WBC count is accurate and can be interpreted as leukopenia. Although the rela-
tive differential values are all within the reference ranges, calculation of absolute counts indicates an absolute neutropenia and lymphopenia. No
unexpected young WBCs are noted, but the morphology indicates hypersegmentation of neutrophils. For the RBCs, the hemoglobin concentration
(8.5) indicates that this person is anemic. Inspection of the MCV indicates macrocytosis, because the MCV is increased to more than 100fL, the
upper limit of the reference range. Examination of the MCHC shows it to be normal, so for these results, the RBC morphology would be described
as macrocytic, normochromic. The elevated RDW indicates substantial anisocytosis. There is no mention of polychromasia, so no young RBCs
are seen. The morphologic description supports the interpretation of the RDW with mention of anisocytosis due to macrocytosis and poikilocytosis
characterized by oval macrocytes and occasional teardrop cells. Howell-Jolly bodies and basophilic stippling are significant RBC inclusions. The
platelet count indicates thrombocytopenia. Although MPV is not reported, the platelets are of normal size and show no morphologic abnormalities,
because there are no notations in the morphologic descriptions.

CBC, Complete blood count; Hb, hemoglobin; Hct, hematocrit; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration; MCV, mean cell volume;
MPV, mean platelet volume; RBC, red blood cell; RDW, RBC distribution width; WBC, white blood cell.

BOX 15-5 Systematic Approach to Complete Blood Count Interpretation

White Blood Cells


Step 1: Ensure that the WBC count is accurate. The presence of nucleated RBCs may require correction of the WBC count.
Step 2: Examine the WBC count for variations in the total number of WBCs; compare the patients count with the laboratorys established reference range.
Steps 3 and 4: Examine the differential information (relative and absolute) on variations in the distribution of WBCs.
Step 5: Make note of immature cells in any cell line reported in the differential that should not appear in normal peripheral blood.
Step 6: Make note of any morphologic abnormalities and correlate film findings with the numerical values.

Red Blood Cells


Step 1: Examine the Hb concentration first to assess anemia.
Step 2: Examine the MCV to assess cell size.
Step 3: Examine the MCHC to assess cell Hb concentration.
Step 4: Examine the RDW to assess anisocytosis.
Step 5: Examine the morphologic description and correlate with the numerical values. Look for evidence of a reticulocyte response.
Step 6: Review remaining information.

Platelets
Step 1: Examine the total platelet count.
Step 2: Examine the MPV to assess platelet size.
Step 3: Examine platelet morphology and correlate with the numerical values.

Hb, Hemoglobin; MCHC, mean cell hemoglobin concentration; MCV, mean cell volume; MPV, mean platelet volume; RBC, red blood cell; RDW, RBC distribution width;
WBC, white blood cell.
208 PART III Routine Laboratory Evaluation of Blood Cells

classify, and the platelets are described simply as bizarre. In Box 15-4 gives an example of how the entire CBC can be
some cases, platelets can be clumped or adherent to WBCs, and summarized using the steps described. When results of the
these arrangements should be noted. As described previously, CBC are properly summarized and no information has been
corrective actions can be taken to derive accurate platelet overlooked, there is confidence that the phase 2 interpretation
and WBC counts when these arrangements are observed on of the results will be reliable. Adopting a methodical approach
the film. to examining each parameter ensures that the myriad informa-
Summarizing platelet parameters includes reporting total tion available from the CBC can be used effectively and effi-
number, platelet size by either instrument MPV or morpho- ciently in patient care. Box 15-5 summarizes the systematic
logic evaluation, and platelet appearance. approach to CBC interpretation.

SUMMARY
Although fewer manual peripheral blood film evaluations are per- The stain used routinely in hematology is Wright or Wright-Giemsa
formed today, much valuable information still can be obtained stain. Staining of all cellular elements occurs when the pH-specific
from a well-made and well-stained film. buffer is added to the stain already on the slide. Staining reactions
The specimen of choice for routine hematology testing is whole depend on the pH of the cells or cellular components.
blood collected in a lavender (purple)topped evacuated tube. The Wright staining is done manually, by automated techniques, or by
tube additive is EDTA, which anticoagulates the blood by chelating quick stains, depending on the venue and the number of slides to
plasma calcium. be processed.
Only rarely does EDTA create problems in analyzing certain indi- Peripheral blood and bone marrow smears always should be evalu-
viduals blood. EDTA-induced platelet clumping or satellitosis ated in a systematic manner, beginning with the 10 objective lens
causes automated analyzers to report falsely decreased platelet and finishing with the 100 oil immersion objective lens. Leukocyte
counts (pseudothrombocytopenia) and falsely increased WBC differential and morphologic evaluation, RBC and platelet morpho-
counts (pseudoleukocytosis). This problem must be recognized logic evaluation, and platelet number estimate all are included.
through blood film examinations and the proper course of action Use of a systematic approach to CBC interpretation ensures that
followed to produce accurate results. all valuable information is assessed and nothing is overlooked.
Numerous methods exist for making peripheral blood films; The systematic approach to CBC interpretation is summarized in
however, the manual wedge film technique is used most Box 15-5.
frequently.
Learning to make consistently good blood films takes practice. Now that you have completed this chapter, go back and
The basic technique can be modified as needed to accommodate read again the case study at the beginning and respond
specimens from patients with very high or very low hematocrits. to the questions presented.

R E V I E W Q UESTIONS
1. A laboratory science student consistently makes wedge- 3. A stained blood film is held up to the light and observed
technique blood films that are too long and thin. What to be bluer than normal. What microscopic abnormality
change in technique would improve the films? might be expected on this film?
a. Increasing the downward pressure on the pusher slide a. Rouleaux
b. Decreasing the acute angle of the pusher slide b. Spherocytosis
c. Placing the drop of blood closer to the center of the c. Reactive lymphocytosis
slide d. Toxic granulation
d. Increasing the acute angle of the pusher slide
4. A laboratorian using the 40 objective lens sees the fol-
2. When a blood film is viewed through the microscope, the lowing numbers of WBCs in 10 fields: 8, 4, 7, 5, 4, 7, 8,
RBCs appear redder than normal, the neutrophils are 6, 4, 6. Which of the following WBC counts most closely
barely visible, and the eosinophils are bright orange. What correlates with the estimate?
is the most likely cause? a. 1.5 109/L
a. The slide was overstained. b. 5.9 109/L
b. The stain was too alkaline. c. 11.8 109/L
c. The buffer was too acidic. d. 24 109/L
d. The slide was not rinsed adequately.
CHAPTER 15 Examination of the Peripheral Blood Film and Correlation with the Complete Blood Count 209

5. A blood film for a very anemic patient with an RBC count 8. Which of the following blood film findings indicates
of 1.25 1012/L shows an average of seven platelets per oil EDTA-induced pseudothrombocytopenia?
immersion field. Which of the following values most a. The platelets are pushed to the feathered end.
closely correlates with the estimate per cubic millimeter b. The platelets are adhering to WBCs.
(or microliter)? c. No platelets at all are seen on the film.
a. 14,000 d. The slide has a bluish discoloration when examined
b. 44,000 macroscopically.
c. 140,000
d. 280,000 9. Using a systematic approach, summarize the WBC para
meters of the CBC presented in the case study at the begin-
6. A blood film for a patient with a normal RBC count has ning of Chapter 34. Use the reference ranges provided
an average of 10 platelets per oil immersion field. Which inside the front cover of this text.
of the following values best correlates with the estimate
per cubic millimeter? 10. Using a systematic approach, summarize the RBC para
a. 20,000 meters of the CBC presented in the case study at the begin-
b. 100,000 ning of Chapter 19.
c. 200,000
d. 400,000

7. What is the absolute count ( 109/L) for the lymphocytes


if the total WBC count is 9.5 109/L and there are 37%
lymphocytes?
a. 3.5
b. 6.5
c. 13
d. 37

REFERENCES
1. Kennedy JB, Maehara KT, Baker AM: Cell and platelet stability platelet gp in a case of EDTA-dependent pseudothrombocy-
in disodium and tripotassium EDTA. Am J Med Technol 47:89- topenia. Am J Clin Pathol 99:163-167, 1993.
93, 1981. 7. Diff-safe blood dispenser. Available at: http://www.
2. Perkins SL: Examination of the blood and bone marrow. In alpha-scientific.com/Diff-safe.html. Accessed December 23,
Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes 2009.
clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer 8. Power KT: The Romanowsky stains: a review. Am J Med Technol
Health/Lippincott Williams & Wilkins, pp 1-20. 48:519-523, 1982.
3. Shahab N, Evans ML: Platelet satellitism. N Engl J Med 9. MacGregor RG, Scott RW, Loh GL: The differential leukocyte
338:591, 1998. count. J Pathol Bacteriol 51:337-368, 1940.
4. Bartels PC, Schoorl M, Lombarts AJ: Screening for EDTA- 10. Briggs C, Harrison P, Machin SJ: Continuing developments
dependent deviations in platelet counts and abnormalities in with the automated platelet count. Int J Lab Hematol 29:77-
platelet distribution histograms in pseudothrombocytope- 91, 2007.
nia. Scand J Clin Lab Invest 57:629-636, 1997. 11. Abe Y, Wada H, Sakakura M, et al: Usefulness of fully auto-
5. Lombarts A, deKieviet W: Recognition and prevention of mated measurement of reticulated platelets using whole
pseudothrombocytopenia and concomitant pseudoleukocy- blood. Clin Appl Thromb Hemost 11:263-270, 2005.
tosis. Am J Clin Pathol 89:634-639, 1988. 12. Ryan DH: Examination of the blood. In Lichtman MA, Beutler
6. De Caterina M, Fratellanza G, Grimaldi E, et al: Evidence E, Kipps TJ, et al, editors: Williams hematology, ed 7, New York,
of a cold immunoglobulin M autoantibody against 78kD 2006, McGraw-Hill, pp 11-19.
16 Bone Marrow Examination
George A. Fritsma*

OUTLINE OBJECTIVES
Bone Marrow Anatomy After completion of this chapter, the reader will be able to:
and Architecture
Indications for Bone 1. Diagram bone marrow architecture and locate 8. List the normal hematopoietic and stromal cells of
Marrow Examination hematopoietic tissue. the bone marrow and their anticipated distribution.
Bone Marrow Specimen 2. List indications for bone marrow examinations. 9. Perform a bone marrow differential count and
Collection Sites 3. Specify sites for bone marrow aspirate and biopsy. compute the myeloid-to-erythroid ratio.
Bone Marrow Aspiration 4. Assemble supplies for performing and assisting in 10. Characterize features of hematopoietic and meta-
and Biopsy bone marrow specimen collection. static tumor cells.
Preparation 5. Assist the physician with bone marrow sample 11. Prepare specimens for and assist in performing spe-
Core Biopsy preparation subsequent to collection. cialized confirmatory bone marrow studies.
Aspiration 6. List the information gained from bone marrow 12. Prepare a systematic written bone marrow examina-
Patient Care
aspirates and biopsy specimens. tion report.
Bone Marrow Sample
7. Perform a bone marrow aspirate smear and core
Management
Direct Aspirate Smears biopsy specimen examination.
Anticoagulated Aspirate
Smears
Crush Smears
Imprints (Touch
CASE STUDY
Preparations)
Concentrate (Buffy Coat) A patient came for treatment complaining of weakness, fatigue, and malaise. Complete blood
Smears count results were as follows:
Histologic Sections (Cell
Hb concentration7.5g/dL
Block)
Marrow Smear Dyes Hct21%
Bone Marrow Aspirate or RBC count2.5 1012/L
Imprint Examination WBC count30 109/L
Low-Power (100) Platelet count540 109/L
Examination Segmented neutrophils21 109/L (70%)
High-Power (500) Immature neutrophils6 109/L (20%)
Examination Basophils1.5 109/L (5%)
Prussian Blue Iron Stain Eosinophils0.3 109/L (1%)
Examination Lymphocytes1.2 109/L (4%)
Bone Marrow Core Biopsy
Bone marrow was hypercellular with 90% myeloid precursors and 10% erythroid precursors.
Specimen Examination
There were 15 megakaryocytes per 10 objective field.
Definitive Bone Marrow
Studies 1. What bone marrow finding provides information on blood cell production?
Bone Marrow 2. What is the myeloid-to-erythroid ratio in this patient, and what does it indicate?
Examination Reports 3. What megakaryocyte distribution is normally seen in a bone marrow aspirate?

*The author thanks Lynne Shaw, MT (ASCP), director of the University of Alabama at Birmingham Hospital Laboratory of Bone Marrow
Pathology, for substantive contribution to this chapter.

210
CHAPTER 16 Bone Marrow Examination 211

fatty metamorphosis increases approximately 10% per decade


BONE MARROW ANATOMY
after 70.4
AND ARCHITECTURE
In adults, bone marrow accounts for 3.4% to 5.9% of body
INDICATIONS FOR BONE
weight, contributes 1600 to 3700g or a volume of 30 to
MARROW EXAMINATION
50mL/kg, and produces roughly six billion blood cells per
kilogram per day in a process called hematopoiesis.1 At birth, Because the procedure is invasive, the decision to collect and
nearly all the bones contain red hematopoietic marrow (see examine a bone marrow specimen requires clinical judgment
Chapter 7). In the fifth to seventh year, adipocytes (fat cells) and the application of inclusion criteria. With the development
begin to replace red marrow in the long bones of the hands, of cytogenetic chromosome studies, flow cytometry, immuno-
feet, legs, and arms, producing yellow marrow, and by late ado- histochemistry, and molecular diagnostics, peripheral blood
lescence hematopoietic marrow is limited to the lower skull, may often provide information historically available from
vertebrae, shoulder, pelvic girdle, ribs, and sternum (see Figure bone marrow only, reducing the demand for marrow speci-
7-2). Although the percentage of bony space devoted to hema- mens. On the other hand, these techniques also augment bone
topoiesis is considerably reduced, the overall volume remains marrowbased diagnosis and thus potentially raise the demand
constant as the individual matures.2 Yellow marrow reverts to for bone marrow examinations in assessment of conditions not
hematopoiesis, increasing red marrow volume, in conditions previously diagnosed through bone marrow examination.
such as chronic blood loss or hemolytic anemia that raise Table 16-1 summarizes indications for examination of bone
demand. marrow.5 Bone marrow examinations may be used to diagnose
The arrangement of red marrow and its relationship to the and stage hematologic and nonhematologic neoplasia, to
central venous sinus is illustrated in Figure 7-3. Hematopoietic determine the cause of cytopenias, and to confirm or exclude
tissue is enmeshed in spongy trabeculae (bony tissue) surround- metabolic and infectious conditions suspected on the basis of
ing a network of sinuses that originate at the endosteum (vas- clinical symptoms and peripheral blood findings.6
cular layer just within the bone) and terminate in collecting Each bone marrow procedure is ordered after consideration
venules.3 Adipocytes occupy approximately 50% of red hema- of clinical and laboratory information. For instance, bone
topoietic marrow space in a 30- to 70-year-old adult, and marrow examination is most likely unnecessary in anemia

TABLE 16-1 Indications for Bone Marrow Examination


Indication Examples
Neoplasia diagnosis Acute leukemias
Myeloproliferative neoplasms such as chronic leukemias, myelofibrosis
Myelodysplastic neoplasms such as refractory anemia
Lymphoproliferative disorders such as acute lymphoblastic leukemia
Immunoglobulin disorders such as plasma cell myeloma, macroglobulinemia
Metastatic tumors
Neoplasia diagnosis and staging Hodgkin and non-Hodgkin lymphoma
Marrow failure: cytopenias Hypoplastic or aplastic anemia
Pure red cell aplasia
Idiosyncratic drug-induced marrow suppression
Myelodysplastic syndromes such as refractory anemia
Marrow necrosis secondary to tumor
Marrow necrosis secondary to severe infection such as parvovirus B19 infection
Immune versus amegakaryocytic thrombocytopenia
Sickle cell crisis
Differentiation of megaloblastic, iron deficiency, sideroblastic, hemolytic, and blood loss anemia
Estimation of storage iron to assess for iron deficiency
Infiltrative processes or fibrosis
Metabolic disorders Gaucher disease
Mast cell disease
Infections Granulomatous disease
Miliary tuberculosis
Fungal infections
Hemophagocytic syndromes
Monitoring of treatment After chemotherapy or radiation therapy to assess minimal residual disease
After stem cell transplantation to assess engraftment
212 PART III Routine Laboratory Evaluation of Blood Cells

Figure 16-1 The posterior superior iliac crest is the favored site for obtaining the bone marrow aspirate and core biopsy specimen because it provides
ample marrow and is isolated from structures that could be damaged by accidental puncture. (Courtesy Indiana Pathology Images, Indianapolis, Ind.).

when the cause is apparent from red blood cell (RBC) indices, lesions. Granulomas, or granulomatous lesions, are cell accu-
serum iron and ferritin levels, or vitamin B12 and folate levels. mulations that contain Langerhans cellslarge, activated granu-
Multilineage abnormalities, circulating blasts in adults, and lar macrophages that look like epithelial cells. Granulomas
unexpected pancytopenia usually prompt marrow examination. signal chronic infection. The biopsy specimen also allows
Bone marrow puncture is prohibited in patients with coagu- morphologic evaluation of bony spicules, which may reveal
lopathies such as hemophilia or vitamin K deficiency, although changes associated with hyperparathyroidism or Paget disease.8
thrombocytopenia is not an absolute contraindication. Special Bone marrow collection sites include the following:
precautions such as bridging therapy may be necessary when a Posterior superior iliac crest (spine) of the pelvis (Figure 16-1).
procedure is performed on a patient receiving antithrombotic In both adults and children, this site provides adequate red
therapy, for instance, warfarin (Coumadin) or heparin. marrow that is isolated from anatomic structures which are
subject to injury. This site is used for both aspiration and
core biopsy.
BONE MARROW SPECIMEN
Anterior superior iliac crest (spine) of the pelvis. This site has
COLLECTION SITES
the same advantages as the posterior superior iliac crest, but
Bone marrow specimen collection is a collaboration between the cortical bone is thicker. This site may be preferred for a
a medical laboratory scientist (medical technologist) and a patient who can only lie supine.
skilled specialty physician, often a pathologist or hematolo- Sternum, below the angle of Lewis at the second intercostal
gist.7 Prior to bone marrow collection, the medical laboratory space. In adults, the sternum provides ample material for
scientist collects peripheral blood for a complete blood count aspiration but is only 1cm thick and cannot be used for
with blood film examination. During collection, the scientist core biopsy. It is possible for the physician to accidentally
assists the physician by managing the specimens and produc- transfix the sternum and enter the pericardium within, dam-
ing initial preparations for examination. aging the heart or great vessels.
Red marrow is gelatinous and amenable to sampling. Most Anterior medial surface of the tibia in children younger than
bone marrow specimens consist of an aspirate (obtained by age 2. This site may be used only for aspiration.
bone marrow aspiration) and a core biopsy specimen (obtained Spinous process of the vertebrae, ribs, or other red marrow
by trephine biopsy), both examined with light microscopy containing bones. These locations are available but are
using 100 and 500 magnification. The aspirate is examined rarely used unless they are the site of a suspicious lesion
to identify the types and proportions of hematologic cells and discovered on a radiograph.
to look for morphologic variance. The core biopsy specimen Adverse outcomes are seen in fewer than 0.05% of marrow
demonstrates bone marrow architecture: the spatial relation- collections. Infections and reactions to anesthetics may occur,
ship of hematologic cells to fat, connective tissue, and bony but the most common side effect is hemorrhage associated
stroma. The core biopsy specimen is also used to estimate with platelet function disorder or thrombocytopenia.
cellularity.
The core biopsy specimen is particularly important for eval-
BONE MARROW ASPIRATION AND BIOPSY
uating diseases that characteristically produce focal lesions,
rather than diffuse involvement of the marrow. Hodgkin lym- Preparation
phoma, non-Hodgkin lymphoma, multiple myeloma, metastatic Less than 24 hours prior to bone marrow collection, the
tumors, amyloid, and granulomas produce predominantly focal medical laboratory scientist collects venous peripheral blood
CHAPTER 16 Bone Marrow Examination 213

for a complete blood count and blood film examination using Vials or test tubes with closures.
a standard collection procedure. Collection is often done Wintrobe hematocrit tubes.
immediately before bone marrow collection. The peripheral Anticoagulant liquid tripotassium ethylenediaminetetraace-
blood specimen is seldom collected after bone marrow collec- tic acid (K3EDTA).
tion to avoid stress-related white blood cell (WBC) count Zenker fixative: potassium dichromate, mercuric chloride,
elevation. sodium sulfate, and glacial acetic acid; B5 fixative: aqueous
Most institutions purchase or assemble disposable sterile mercuric chloride and sodium acetate, or 10% neutral for-
bone marrow specimen collection trays that provide the malin. Because Zenker fixative and B5 contain toxic mercury,
following: controlled disposal is required.
Surgical gloves. Gauze dressings.
Shaving equipment. The patient is asked to lie supine, prone, or in the right or
Antiseptic solution and alcohol pads. left lateral decubitus position (lying on the right or left side).
Drape material. With attention to standard precautions, the skin is shaved if
Local anesthetic injection, usually 1% lidocaine, not to
exceed 20mL per patient.
No. 11 scalpel blade for skin incision.
Disposable Jamshidi biopsy needle (Care Fusion; Figure Puncture needle
16-2) or Westerman-Jensen needle (Becton, Dickinson and with obturator
Company, Franklin Lakes, N.J.; Figure 16-3). Both provide
an obturator, core biopsy tool, and stylet. A Snarecoil biopsy
needle also is available (Kendall Company, Mansfield,
Mass.). The Snarecoil has a coil mechanism at the needle
tip that allows for capture of the bone marrow specimen Core biopsy
needle with
without needle redirection (Figure 16-4). expulsion stylus
Disposable 14- to 18-gauge aspiration needle with obtura-
Figure 16-2 Disposable sterile Jamshidi bone marrow biopsy and aspira-
tor. Alternatively, the University of Illinois aspiration needle
tion needle. The outer puncture cannula is advanced to the medulla with the
may be used for sternal puncture. The University of Illinois obturator in place to prevent bone coring. The physician removes the obtura-
needle provides a flange that prevents penetration of the tor and slides the core biopsy needle through the cannula and into the
sternum to the pericardium. medulla with the expulsion stylus removed. The core biopsy needle is
Microscope slides or coverslips washed in 70% ethanol. removed from the puncture needle with the specimen in place. The specimen
Petri dishes or shallow circular watch glasses. is expelled using the stylus. (Courtesy Care Fusion, McGaw Park, Ill.)

Biopsy needle Obturator Cutting blade

Stylet

2
Remove obturator
Puncture through cortex Insert cutting blades
1
(obturator in needle)
Partially withdraw cutting
blades with specimen while
needle remains fixed

Twist and remove needle


3
and cutting blades together

Figure 16-3 Bone marrow biopsy technique using the Westerman-Jensen needle.
214 PART III Routine Laboratory Evaluation of Blood Cells

cavity attachments, and the biopsy needle and cannula are


withdrawn from the bone, taking the core cylinder with them.
The core cylinder is 1 to 1.5cm long and 1 to 2mm in diam-
eter, and weighs about 150mg. The biopsy needle is placed
over an ethanol-cleaned slide and the stylus is pushed through
to dislodge the core cylinder. Using sterile forceps, the scientist
prepares imprints (touch preparations) and transfers the core
cylinder to the chosen fixative, Zenker, B5, or formalin.
When the Westerman-Jensen needle is used, the obturator
is removed, the cutting blades are inserted through the cannula,
and the blades are advanced into the medullary cavity. The
cutting blades are pressed into the medullary bone, with the
outer cannula held firmly in a stationary position. The blades
are withdrawn so that the cannula entraps the tissue, and the
entire unit is withdrawn. The core cylinder is removed by
inserting the probe through the cutting tip and extruding the
specimen through the hub of the needle to the selected slide
and fixative-containing receptacles.

Aspiration
In a separate location from the biopsy, a 14- to 18-gauge aspira-
tion needle such as the University of Illinois needle, with obtu-
rator, is inserted through the skin and cortex of the bone. The
obturator is removed and a 10- to 20-mL syringe is attached.
The plunger is withdrawn to create negative pressure and aspi-
rate 1.0 to 1.5mL of marrow into the syringe. Collecting more
Figure 16-4 Snarecoil bone marrow biopsy needle. The coil mechanism
than 1.5mL dilutes the hematopoietic marrow with sinusoidal
resides within the biopsy needle as illustrated in the magnified image. The
coil is turned to draw the marrow specimen into the needle. (Courtesy Tyco (peripheral) blood. The syringe is detached and is passed imme-
Healthcare/Kendall, Mansfield, Mass.) diately to the scientist, who expels the material onto a series of
clean and sterile microscopic slides or coverslips. A second
syringe may be attached and additional specimen aspirated for
necessary, disinfected, and draped. The skin, dermis, and sub- cytogenetic analysis, molecular diagnosis (polymerase chain reaction
cutaneous tissue are infiltrated with a local anesthetic solution, testing), or immunophenotyping using flow cytometry. The needle
such as 1% or 2% lidocaine or procaine, through a 25-gauge is then withdrawn, and pressure is applied to the wound.
needle, producing a 0.5- to 1.0-cm papule (bubble). The If no marrow is obtained, the physician returns the obtura-
25-gauge needle is replaced with a 21-gauge needle, which is tor to the needle, advances the needle, attaches a fresh syringe,
inserted through the papule to the periosteum (bone surface). and tries again. The syringe and needle are retracted slightly
With the point of the needle on the periosteum, approximately and the process is repeated. If this attempt is unsuccessful, the
2mL of anesthetic is injected over a dime-sized area as the needle and syringe are removed, pressure is applied, and the
needle is rotated, after which the anesthesia needle is with- procedure is begun at a new site. If the marrow is fibrotic, acel-
drawn. Next, a 3-mm skin incision is made over the puncture lular, or packed with leukemic cells, the aspiration may be
site with a No. 11 scalpel blade to prevent skin coring during unsuccessful, known as a dry tap. In this case, a biopsy is neces-
insertion of the needle. sary, and cell morphology may be observed using a slide
imprint, or touch preparation.
Core Biopsy
The biopsy specimen is usually collected first, because aspira- Patient Care
tion may destroy marrow architecture. After the incision is Subsequent to bone marrow biopsy or aspiration, a pressure
made, the Jamshidi outer cannula with the obturator in place dressing is applied, and the patient is required to remain in the
is inserted through the skin and cortex of the bone. The obtura- same position for 60 minutes to prevent bleeding.
tor prevents coring of skin or bone. Reciprocating rotation
promotes the forward advancement of the cannula until the
BONE MARROW SAMPLE MANAGEMENT
resistance weakens, which indicates penetration through the
cortex to the medullary cavity of the bone. The obturator is Direct Aspirate Smears
removed and the biopsy needle is inserted through the cannula The medical laboratory scientist receives the aspirate syringe
and advanced slowly 2 to 3cm with continued reciprocating from the physician at the bedside and immediately transfers
rotation along the long axis. The needle angle is changed drops of the marrow specimen onto six to eight ethanol-
slightly to separate the core cylinder specimen from its marrow washed microscope slides. Marrow clots rapidly, so good
CHAPTER 16 Bone Marrow Examination 215
organization is essential. Using spreader slides, the scientist of imprint preparations may imitate aspirate morphology,
spreads the drop into a wedge-shaped smear 1 2 to 3 4 the length although few spicules are transferred, and imprints are valuable
of the slide, similar to a peripheral blood film. Bony spicules when the specimen has clotted or there is a dry tap.
0.5 to 1.0mm in diameter and larger fat globules follow
behind the spreader and deposit on the slide. In the direct Concentrate (Buffy Coat) Smears
smear preparation, the spicules are not squashed. The smears Buffy coat smears are useful when there are sparse nucleated
may be fanned to promote rapid drying in an effort to preserve cells in the direct marrow smear or when the number of nucle-
cell morphology. ated cells is anticipated to be small, as in aplastic anemia.
In the syringe, the specimen is largely peripheral blood with Approximately 1.5mL of K3EDTA-anticoagulated marrow
suspended light-colored bony spicules and fat globules. The specimen is transferred to a narrow-bore glass or plastic tube
scientist evaluates the blood for spicules: more spicules mean such as a Wintrobe hematocrit tube. The tube is centrifuged at
a specimen with more cells to identify and categorize. If the 2500g for 10 minutes and examined for four layers.
specimen has few fat globules or spicules the scientist may alert The top layer is yellowish fat and normally occupies 1% to
the physician to collect an additional specimen. 3% of the column. The second layer, plasma, varies in volume
depending on the amount of peripheral blood in the specimen.
Anticoagulated Aspirate Smears The third layer consists of nucleated cells and is called the
Anticoagulated specimens are a more leisurely alternative to myeloid-erythroid (ME) layer. The ME layer is normally 5% to
direct aspirate smears. The aspirate is expressed from the 8% of the total column. The bottom layer is RBCs, and its
syringe into a vial of K3EDTA and subsequently pipetted to volume, like that of the plasma layer, depends on the amount
clean glass slides and spread using the same approach as in of peripheral blood present. The scientist records the ratio of
direct aspirate smear preparation. All anticoagulants distort cell the fat and ME layers using millimeter gradations on the tube.
morphology; however, K3EDTA generates the least distortion. Once the column is examined, the scientist aspirates a
portion of the ME layer with a portion of plasma and transfers
Crush Smears the suspension to a Petri dish or watch glass. Marrow smears
To prepare crush smears, the medical laboratory scientist expels are subsequently prepared using the crush smear technique.
a portion of the aspirate to a Petri dish or watch glass covered The concentrated buffy coat smear compensates for hypo-
with a few milliliters of K3EDTA solution and spreads the aspi- cellular marrow and allows for examination of large numbers
rate over the surface with a sterile applicator. Individual bony of nucleated cells without interference from fat or RBCs. On
spicules are transferred using applicators, forceps, or micropi- the other hand, cell distribution is distorted by the procedure.
pettes (preferred) to several ethanol-washed glass slides. Addi- Therefore, the scientist does not estimate numbers of different
tional glass slides are placed directly over the specimens at right cell types or maturation stages on a buffy coat smear.
angles and pressed gently to crush the spicules. The slides are
separated laterally to create two rectangular smears, which may Histologic Sections (Cell Block)
be fanned to encourage rapid drying. After aspirate smears are prepared and aliquots of marrow have
Some scientists prefer to transfer aspirate directly to the been distributed for cytogenetic, molecular, and immuno
slide, subsequently tilting the slide to drain off peripheral phenotypic studies, the remaining core biopsy specimen,
blood while retaining spicules. Once drained, the spicules are spicules, or clotted specimen is submitted for histologic exami-
then crushed with a second slide as described earlier. nation. The specimen is suspended in 10% formalin, Zenker
The scientist may add one drop of 22% albumin to the glacial acetic acid, or B5 fixative for approximately 2 hours.
EDTA solution, particularly if the specimen is suspected to The fixed specimen is centrifuged, and the pellet is decalci-
contain prolymphocytes or lymphoblasts, which tend to fied and wrapped in an embedding bag or lens paper and
rupture. The albumin reduces the occurrence of smudge or placed in a paraffin embedding cassette. The embedded speci-
basket cells often seen in lymphoid marrow lesions. men is sectioned and dyed with hematoxylin and eosin (H&E)
The crush preparation procedure may also be performed dye and examined.
using ethanol-washed coverslips in place of slides. The cover-
slip method demands adroit manipulation but may yield Marrow Smear Dyes
better morphologic information, because the smaller cover- Marrow aspirate smears are stained with Wright or Wright-
slips generate less cell rupture during separation. Use of glass Giemsa dyes using the same protocols as for peripheral blood
slides offers the opportunity for automated staining, whereas film staining. Some laboratory managers increase staining time
coverslip preparations must first be affixed smear side up to to compensate for the relative thickness of marrow smears
slides and then stained manually (see Chapter 15). compared with peripheral blood films.
Marrow aspirate smears and core biopsy specimens may
Imprints (Touch Preparations) also be stained using a ferric ferricyanide (Prussian blue) solu-
Core biopsy specimens and clotted marrow may be held in tion to detect and estimate marrow storage iron or iron meta
forceps and repeatedly touched to a washed glass slide or bolism abnormalities (see Chapter 19). Further, a number of
coverslip, so that cells attach and rapidly dry. The scientist lifts cytochemical dyes are used for cell identification or differentia-
directly upward to prevent cell distortion. The cell morphology tion (Table 16-2 and Chapter 30).
216 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 16-2 Cytochemical Dyes Used to Identify Bone


Marrow Cells and Maturation Stages
Cytochemical Dye Application
Myeloperoxidase (MPO) Detects myelocytic cells by staining
cytoplasmic granular contents
Sudan black B (SBB) Detects myelocytic cells by staining
cytoplasmic granular contents
Periodic acidSchiff Detects lymphocytic cells and certain
(PAS) abnormal erythrocytic cells by staining
of cytoplasmic glycogen
Esterases Distinguish myelocytic from monocytic
maturation stages (several esterase
substrates)
Tartrate-resistant acid Detects tartrate-resistant acid phosphatase Figure 16-5 Bone marrow aspirate specimen illustrating intact, contigu-
phosphatase (TRAP) granules in hairy cell leukemia ous cells and adipocytes. This is a good site for morphologic evaluation and
cell counting (Wright stain, 100).

for examination, whereas areas showing distorted morphology


BOX 16-1 Bone Marrow Aspirate Microscopic Smear
or dilution with sinusoidal blood are avoided.
Examination: Low and High Power
Near the spicules, cellularity is estimated by observing the
proportion of hematopoietic cells to adipocytes (clear fat
Low Power: 10 Objective
areas).9 For iliac crest marrow, 50% cellularity is normal for
Assess peripheral blood dilution
patients aged 30 to 70 years. In childhood, cellularity is 80%,
Find bony spicules and areas of clear cell morphology
and after age 70, cellularity is reduced. For those older than age
Observe fat-to-marrow ratio, estimate cellularity
70, a rule of thumb is to subtract patient age from 100% and
Search for tumor cells in clusters
add 10%. Thus for a 75-year-old, the anticipated cellularity is
Examine and estimate megakaryocytes
15% to 35%. By comparing with the age-related normal cel-
lularity values, the microscopist classifies the observed area as
High Power: 50 and 100 objective
hypocellular, normocellular, or hypercellular. If a core biopsy speci-
Observe myelocytic and erythrocytic maturation
men was collected it provides a more accurate estimate of
Distinguish abnormal distribution of cells or cell maturation stages
cellularity than an aspirate smear, because in aspirates there is
Perform differential count on 300 to 1000 cells
always some dilution of hematopoietic tissue with sinus blood.
Compute myeloid-to-erythroid ratio
In the absence of leukemia, lymphocytes should total fewer
than 30% of nucleated cells; if more are present, the marrow
specimen has been substantially diluted and should not be
used to estimate cellularity.10
BONE MARROW ASPIRATE OR
Using the 10 objective the microscopist searches for abnor-
IMPRINT EXAMINATION
mal, often molded, cell clusters (syncytia) of metastatic tumor
Box 16-1 describes the uses of low- and high-power objectives cells or lymphoblasts. Tumor cell nuclei often stain darkly (hyper-
in examining bone marrow aspirate direct smears or imprints. chromatic), and vacuoles are seen in the cytoplasm. Tumor cell
clusters are often found near the edges of the smear.
Low-Power (100 ) Examination Although myelocytic cells and erythrocytic cells are best
Once the bone marrow aspirate direct smear or imprint is examined using 500 magnification, they may be more easily
prepared and stained, the scientist or pathologist begins the distinguished using the 10 objective. The erythrocytic matura-
microscopic examination using the low-power (10) dry lens, tion stages stain more intensely and their margins are more
which, when coupled with 10 oculars, provides a total 100 sharply defined, features more easily distinguished at lower
magnification. Most bone marrow examinations are performed magnification.
using a teaching report format that employs projection or The microscopist evaluates megakaryocytes using low power
multiheaded microscopes to allow viewing by residents, (Figure 16-6). Megakaryocytes are the largest cells in the bone
fellows, medical laboratory science students, and attending marrow, 30 to 50m in diameter, with multilobed nuclei (see
staff. The microscopist locates the bony spicules, aggregations Chapter 13). Although in special circumstances microscopists
of bone and hematopoietic marrow, which stain dark blue may differentiate three megakaryocyte maturation stages
(Figure 16-5). In imprints, spicules are sparse or absent, and megakaryoblast, promegakaryocyte, and megakaryocyte (MK-I to
the search is for hematopoietic cells instead. Within these MK-III)a total megakaryocyte estimate is generally satis
areas, intact and nearly contiguous nucleated cells are selected factory. In a well-prepared aspirate or biopsy specimen, the
CHAPTER 16 Bone Marrow Examination 217

MyBl
Myel

OrthoN
Meta
ProMy
Myel
Lymph

ProMy
ProMy

Figure 16-8 Bone marrow aspirate smear. Myelocytic stages include a


myeloblast (MyBl), promyelocytes (ProMy), a myelocyte (Myel), and meta
myelocyte (Meta). One orthochromic normoblast (OrthoN) and one lympho-
cyte (Lymph) are present (Wright stain, 1000).
Figure 16-6 Bone marrow aspirate smear showing megakaryocyte with
budding platelets at the plasma membrane (Wright stain, 1000). Mega-
karyocytes are counted at 100 magnification, but if there is abnormal
morphology, cells are examined at 500 or 1000.
Meta Band

ProMy
Myel

Myel Band

Myel Myel
MyBl

Figure 16-9 Bone marrow aspirate smear. Myelocytic stages include


Lymph
myelocytes (Myel), a metamyelocyte (Meta), and neutrophilic bands (Wright
stain, 1000).
Figure 16-7 Bone marrow aspirate smear. Myelocytic stages include a
myeloblast (MyBl), promyelocyte (ProMy), and myelocyte (Myel). The lympho-
cyte (Lymph) diameter illustrates its size relative to the myelocytic stages. SEG
The source of the lymphocyte is sinus blood (Wright stain, 1000). SEG

Band
microscopist observes 2 to 10 megakaryocytes per low-power
SEG
field. Deviations yield important information reported as
decreased or increased megakaryocytes. Bone marrow mega- Band

karyocyte estimates are essential to the evaluation of peripheral SEG


Band
blood thrombocytopenia and thrombocytosis; for instance, in
SEG
immune thrombocytopenia, marrow megakaryocytes prolifer-
ate markedly. Band
Abnormal megakaryocytes may be small, lack granularity,
or have poorly lobulated or hyperlobulated nuclei. Indications Figure 16-10 Bone marrow aspirate smear illustrating neutrophilic bands
and segmented neutrophils (SEGs) (Wright stain, 1000).
of abnormality may be visible using low power; however,
conclusive descriptions require 500 or even 1000 total
magnification. eosinophils, basophils, lymphocytes, plasma cells, monocytes,
and histiocytes. See Chapters 7, 8, and 12 for detailed cell and
High-Power (500 ) Examination stage descriptions. Table 16-3 names all normal marrow cells
Having located a suitable examination area, the microscopist and provides their expected percentages.
places a drop of immersion oil on the specimen and switches The microscopist searches for maturation gaps, misdistribu-
to the 50 objective, providing 500 total magnification. All tion of maturation stages, and abnormal morphology. Although
of the nucleated cells are reviewed for morphology and normal the specimen is customarily reviewed using the 50 oil immer-
maturation. Besides megakaryocytes, cells of the myelocytic sion objective, the 100 oil immersion objective is frequently
(Figures 16-7 through 16-10) and erythrocytic (rubricytic, nor- employed to detect small but significant morphologic abnor-
moblastic, Figure 16-11) series should be present, along with malities in the nuclei and cytoplasm of suspect cells.
218 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 16-3 Anticipated Distribution of Cells and Cell Maturation Stages in Aspirates or Imprints
Cell or Cell Maturation Stage Distribution Cell or Cell Maturation Stage Distribution
Myeloblasts 0-3% Pronormoblasts/rubriblasts 0-1%
Promyelocytes 1-5% Basophilic normoblasts/prorubricytes 1-4%
Myelocytes 6-17% Polychromatophilic normoblasts/rubricytes 10-20%
Metamyelocytes 3-20% Orthochromic normoblasts/metarubricytes 6-10%
Neutrophilic bands 9-32% Lymphocytes 5-18%
Segmented neutrophils 7-30% Plasma cells 0-1%
Eosinophils and eosinophilic precursors 0-3% Monocytes 0-1%
Basophils and mast cells 0-1% Histiocytes 0-1%
Megakaryocytes 2 to 10 visible per low-power field Myeloid-to-erythroid ratio 1.5:1-3.3:1
: :

Figure 16-11 Bone marrow aspirate smear showing an island of eryth- Figure 16-12 Bone marrow aspirate smear showing a cluster of osteo-
rocytic precursors with polychromatophilic and orthochromic normoblasts blasts that superficially resemble plasma cells. Osteoblasts have round to
(Wright stain, 1000). oval eccentric nuclei and mottled blue cytoplasm that is devoid of secretory
granules. They may have a clear area within the cytoplasm, but lack the
Many laboratory directors require a differential count of well-defined central Golgi complex of the plasma cell (Wright stain, 1000).
300 to 1000 nucleated cells. These seemingly large totals are
rapidly reached in a well-prepared bone marrow smear at 500 cells. Osteoblasts are usually found in clusters resembling
magnification and compensate statistically for the anticipated myeloma cell clusters. Their presence in marrow aspirates and
uneven distribution of spicules and hematopoietic cells. The core biopsy specimens is incidental; they do not signal disease,
microscopist counts cells and maturation stages surrounding but they may create confusion.
several spicules to maximize the opportunity for detecting Osteoclasts are nearly the diameter of megakaryocytes, but
disease-related cells. Some laboratory directors eschew the their multiple, evenly spaced nuclei distinguish them from
differential in favor of a thorough examination of the smear. multilobed megakaryocyte nuclei (Figure 16-13). Osteoclasts
Many microscopists choose not to differentiate the four appear to derive from myeloid progenitor cells and are respon-
nucleated erythrocytic maturation stages, and others may sible for bone resorption, acting in concert with osteoblasts.
combine three of the fourbasophilic, polychromatophilic, and Osteoclasts are recognized more often in core biopsy speci-
orthochromic normoblastsin a single total, counting only pro- mens than in aspirates.
normoblasts separately. In normal marrow, most erythrocytic Adipocytes, endothelial cells that line blood vessels, and
precursors are either polychromatophilic or orthochromic nor- fibroblast-like reticular cells complete the bone marrow stroma
moblasts, and differentiation yields little additional informa- (see Chapter 7). Stromal cells and their extracellular matrix
tion. On the other hand, differentiation may be helpful in provide the suitable microenvironment for the maturation and
megaloblastic, iron deficiency or refractory anemias. proliferation of hematopoietic cells, but are seldom examined
The microscopist may infrequently find osteoblasts and osteo- for diagnosis of hematologic or systemic disease.
clasts (Figure 16-12). Osteoblasts are responsible for bone for- Finally, Langerhans cells, giant cells with palisade nuclei
mation and remodeling, and derive from endosteal (inner found in granulomas, signal chronic inflammation.
lining) cells. Osteoblasts resemble plasma cells with eccentric Once the differential is completed, the myeloid-to-erythroid
round to oval nuclei and abundant blue, mottled cytoplasm, (M:E) ratio is computed from the total of myeloid to the total
but lack the prominent Golgi apparatus characteristic of plasma of nucleated erythroid cell stages. Excluded from the M:E ratio
CHAPTER 16 Bone Marrow Examination 219

are lymphocytes, plasma cells, monocytes, histiocytes, non- The iron stain may be used for core biopsy specimens, but
nucleated erythrocytes, and nonhematopoietic stromal cells. decalcifying agents used to soften the biopsy specimen during
processing may leech iron, which gives a false impression of
Prussian Blue Iron Stain Examination decreased or absent iron stores. For this reason, the aspirate is
A Prussian blue (ferric ferricyanide) iron stain is commonly favored for the iron stain if sufficient spicules are present.
used on the aspirate smear. Figure 16-14 illustrates normal iron,
absence of iron, and increased iron stores in aspirate smears.
BONE MARROW CORE BIOPSY
SPECIMEN EXAMINATION
The standard dye for the core biopsy specimen is H&E. Other
dyes and their purposes are listed in Table 16-4.
Bone marrow core biopsy specimen and imprint (touch
preparation) examinations are essential when the aspiration
procedure yields a dry tap, which may be the result of hypo-
plastic or aplastic anemia, fibrosis, or tight packing of the
marrow cavity with leukemic cells. The key advantage of the
core biopsy specimen is preservation of bone marrow architec-
ture so that cells, tumor clusters (Figure 16-15), and matura-
tion stages may be examined relative to stromal elements. The
disadvantage is that individual hematopoietic cell morphology
is obscured.
The microscopist first examines the core biopsy specimen
Figure 16-13 Bone marrow core biopsy section from a patient with preparation using the 10 objective (100 total magnification)
hyperparathyroidism. The large multinucleated cell near the endosteal to assess cellularity. Because the sample is larger, the core biopsy
surface is an osteoclast, a cell that reabsorbs bone. The spindle-shaped cells specimen provides a more accurate estimate of cellularity than
are fibroblasts (hematoxylin and eosin stain, 500). the aspirate. The estimate is made by comparing cellular areas

A B

C
Figure 16-14 Prussian blue dye on bone marrow aspirate smears (500). A, Normal iron stores. B, Absence of iron stores. C, Increased iron stores.
220 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 16-4 Dyes Used in Examination of Bone Marrow volume, megakaryocyte numbers are assessed more accurately
Core Biopsy Specimens by examining a core biopsy section than an aspirate smear.
Normally there are 2 to 10 megakaryocytes per 10 field, the
Dye Comment
same as in an aspirate smear or imprint.
Hematoxylin and Used to evaluate cellularity and hematopoietic Using the 50 oil immersion objective, the microscopist
eosin (H&E) cell distribution, locate abnormal cell next observes cell distribution relative to bone marrow stroma.
clusters. For instance, in people older than age 70, normal lymphocytes
Prussian blue (ferric Used to evaluate iron stores for deficiency or may form small aggregates in nonparatrabecular regions,
ferricyanide) iron excess iron. Decalcification may remove whereas malignant lymphoma cell clusters are often paratrabecu-
stain iron from fixed specimens; thus lar. In addition, normal lymphocytes remain as discrete cells,
ethylenediaminetetraacetic acid chelation or whereas lymphoma cells are pleomorphic and syncytial.
the aspirate smear is preferred for iron If no aspirate or imprint smears could be prepared, the core
store estimation. biopsy specimen may be stained using Wright, Giemsa, or
Reticulin and Used to examine for marrow fibrosis. Wright-Giemsa dyes to make limited observations of cellular
trichrome dyes morphology. In Wright- or Giemsa-stained biopsy sections,
Acid-fast stains Used to examine for acid-fast bacilli, fungi, or myeloblasts and promyelocytes have oval or round nuclei with
bacteria in granulomatous disease. cytoplasm that stains blue (Figure 16-18). Neutrophilic myelo-
Gram stain Used to examine for acid-fast bacilli, fungi, or cytes and metamyelocytes have light pink cytoplasm. Mature
bacteria in granulomatous disease. segmented neutrophils (SEGs) and neutrophilic bands are rec-
Immunohistochemical Dye-tagged monoclonal antibodies specific for ognized by their smaller diameter and darkly stained C-shaped
dyes tumor surface markers are used to nuclei (bands) or nuclear segments (SEGs). The cytoplasm of
establish the identity of malignant cells. bands and SEGs may be light pink or may seem unstained
Wright or Wright- Used to observe hematopoietic cell structure. (Figure 16-19).
Giemsa dyes Cell identification is less accurate in a The cytoplasm of eosinophils stains red or orange, which
biopsy specimen than in an aspirate smear. makes them the most brightly stained cells of the marrow.
Basophils cannot be recognized on marrow biopsy specimens
fixed with Zenker glacial acetic acid solution.
The microscopist may find it difficult to differentiate myelo-
cytic cells from erythrocytic cells in biopsy specimens other
than to observe that the latter tend to cluster with more mature
normoblasts and often surround histiocytes. Polychromato-
philic and orthochromic normoblasts, the two most common
erythrocytic maturation stages, have centrally placed, round
nuclei that stain intensely (Figure 16-20). Their cytoplasm is
not appreciably stained, but the plasma membrane margin is
clearly discerned, which gives the cells of these stages a fried
egg appearance. Because erythrocytic cells have a tendency
to cluster in small groups, they are more easily recognized
using the 10 objective, although their individual morphology
cannot be seen.
Lymphocytes are among the most difficult cells to recognize
Figure 16-15 Bone marrow aspirate smear showing a cluster or syncytia in the core biopsy specimen, unless they occur in clusters.
of tumor cells. Nuclei are irregular and hyperchromatic, and cytoplasm is Mature lymphocytes exhibit speckled nuclear chromatin in a
vacuolated. Cytoplasmic margins are poorly delineated (Wright stain, 500). small, round nucleus, along with a scant amount of blue cyto-
plasm (Figure 16-21). Immature lymphocytes (prolympho-
with the clear-appearing adipocytes, using a method identical to cytes) have larger round or lobulated nuclei but still only a
that employed in examination of aspirate smears. All fields are small rim of blue cytoplasm.
examined, because cells distribute unevenly. Examples of hypo- Plasma cells are difficult to distinguish from myelocytes in
cellular and hypercellular core biopsy sections are provided for H&E-stained sections but are recognized using Wright-Giemsa
comparison with normocellular marrow in Figure 16-16. dye as cells with eccentric dark nuclei and blue cytoplasm and
Megakaryocytes are easily recognized by their outsized a prominent pale central Golgi apparatus (Figure 16-22). Char-
diameter and even distribution throughout the biopsy. They acteristically, plasma cells are located adjacent to blood vessels.
exhibit the characteristic lobulated nucleus, although nuclei of
the more mature megakaryocytes are smaller and more darkly
DEFINITIVE BONE MARROW STUDIES
stained in H&E preparations than on a Wright-stained aspirate.
Their cytoplasm varies from light pink in younger cells to dark Although in many cases the aspirate smear and biopsy
pink in older cells (Figure 16-17). Owing to the greater sample specimen are diagnostic, additional studies may be needed.
CHAPTER 16 Bone Marrow Examination 221

A B

C
Figure 16-16 A, Representative core biopsy section showing normal cellularity, approximately 50% fat and 50% hematopoietic cells (hematoxylin
and eosin, 50). B, Hypocellular core biopsy specimen with only fat and connective tissue cells from a patient with aplastic anemia (hematoxylin and eosin
stain, 100). C, Hypercellular core biopsy specimen from a patient with chronic myelogenous leukemia. There is virtually 100% cellularity with no fat visible
(hematoxylin and eosin stain, 100). (B courtesy Dennis P. OMalley, MD, director, Immunohistochemistry Laboratory, Indiana University School of Medicine,
Indianapolis, IN.)

Figure 16-17 Core biopsy section containing many large lobulated mega- Figure 16-18 Core biopsy section infiltrated by blasts with blue cyto-
karyocytes and increased blasts (Giemsa stain, 400). plasm and a few myelocytes with pink cytoplasm (Giemsa, 400).

Such studies and their applications are given in Table 16-5.


BONE MARROW EXAMINATION REPORTS
These studies require additional bone marrow volume and
specialized specimen collection. Information on Prussian blue The components of a bone marrow report should be generated
iron stains and cytochemistry was provided earlier. Each study systematically and are given in Table 16-6. An example of a
is described in the chapter referenced in the Table 16-5. bone marrow examination report is provided in Figure 16-23.
222 PART III Routine Laboratory Evaluation of Blood Cells

Figure 16-19 Core biopsy section showing myelocytes, metamyelocytes, Figure 16-20 Erythrocytic island in a core biopsy section. Late-stage
bands, segmented neutrophils, and bright red-orange eosinophils (Giemsa normoblasts often have a fried egg appearance (Giemsa stain, 400).
stain, 400).

Figure 16-21 Core biopsy section showing small mature lymphocytes. A Figure 16-22 Plasma cells in a core biopsy section. Nuclei are eccentric,
few are immature with larger nuclei containing a single prominent nucleolus cytoplasm is blue with a prominent central unstained Golgi apparatus (Giemsa
(Giemsa, 400). stain, 400).

TABLE 16-5 Definitive Studies Performed on Selected Bone Marrow Specimens


Bone Marrow Study Application Specimen Chapter
Iron stain Identification of iron deficiency, iron overload Fresh marrow aspirate 19
Cytochemical studies Diagnosis of leukemias and lymphomas Fresh marrow aspirate 30
Cytogenetic studies Diagnosis of acute leukemias via deletions, 1mL marrow in heparin 31
translocations, and polysomy; remission studies
Molecular studies Polymerase chain reaction for diagnostic point mutations; 1mL marrow in EDTA 32
minimal residual disease studies
Fluorescent in situ hybridization Staining for diagnostic mutations; minimal residual Fresh marrow aspirate 32
disease studies
Flow cytometry Immunophenotyping, usually of malignant hematopoietic 1mL marrow in heparin, 33
cells, clonality; minimal residual disease studies EDTA, or ACD

ACD, Acid-citrate dextrose; EDTA, ethylenediaminetetraacetic acid.


CHAPTER 16 Bone Marrow Examination 223

Laboratory of Bone Marrow Pathology Report


Name Room number and Unit Patient number Specimen number

Date Age Sex Race

Address

City, State, Zip

Site Aspirate Biopsy Markers Cytogenetics and Molecular Studies

Attending Physician Pathologist

Clinical Diagnosis Pathology Diagnosis

Acute leukemia Acute myelomonocytic leukemia (FAB M4) with


mild eosinophilia. M98613, M47150, D 40800

Differential
Date PB Marrow Marrow PB Marrow Marrow
Range Range
(percent) (percent)

Blast, unclassified 4 77 0-2 Normal lymphocytes 50 2 3-24

Myeloblasts 3-5 Reactive lymphocytes

Promyelocytes 1-8 Lymphoblasts

Myelocytes 4 5-21 Prolymphocytes

Metamyelocytes 6-22 Monocytes 3 0-3

Band neutrophils 6-22 Plasma cells 0-2

Segmented neutrophils 40 4 9-27 Rubriblasts 0-4

Eosinophils 3 4 Prorubricytes 1-6

Basophils Rubricytes 5-25

Other Metarubricytes 1-21

Other Erythroid 8 10-30

WBC PLT MPV HGB HCT MCHC


2,300 89,000 8 8.1 23 110
Peripheral blood film: Leukopenia, few blasts, myelocytes, neutrophils. Relative lymphocytosis. RBCs normochromic,
macrocytes, mild anisocytosis. Rare teardrop cells. Normal platelet morphology.
Bone marrow aspirate: Few spicules, blasts 70-90% with large nuclei with fine chromatin and delicate creases. Moderate gray
cytoplasm. Few blasts with high N/C ratio and large cells with cytoplasmic blebs. Few mature myeloid precursors. Erythroid
precursors markedly decreased, with rare binucleate rubricytes. Few eosinophil precursors noted. Dysmorphic megakaryocytes
with increased nuclear lobulation.
Bone marrow imprint: Significant blast increase, most have myelomonocytic morphology. Few mature myeloid precursors,
erythroid precursors markedly decreased.
Bone marrow biopsy: Hypercellular, 50-80%. Most cells are blasts, few mature myeloid precursors. Erythroid and megakaryocytic
precursors markedly reduced.
Iron stain: Stainable iron in macrophages. Sideroblasts and rare ringed sideroblasts.
Flow cytometry: Bright CD15, HLA-DR, CD33, CD56, and CD11c. CD14 minimally expressed. Bright CD15 and CD11c consistent
with monocytic differentiation.

AMML with eosinophilia (FAB M4e) cannot be ruled out, chromosome analysis pending. Rare ringed sideroblasts
suggest transformation from preexisting myelodysplastic syndrome.

Figure 16-23 Example of a bone marrow examination report, including reference intervals, peripheral blood complete blood count results, marrow
differential, and narrative. PB, Peripheral blood; BMA, bone marrow aspirate; BMBX, bone marrow biopsy specimen; DX, diagnosis.
224 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 16-6 Components of a Bone Marrow Examination Report


Component Description
Patient history Patient identity and age, narration of symptoms, physical findings, findings in kindred, treatment
Complete blood count (CBC) Peripheral blood CBC collected no more than 24 hours before the bone marrow puncture, includes hemogram
and peripheral blood film examination
Cellularity Hypocellular, normocellular, or hypercellular classification based on ratio of hematopoietic cells to adipocytes
Megakaryocytes Estimate at 10 magnification compared with reference interval and comment on morphology
Maturation Narrative characterizing the maturation of the myelocytic and erythrocytic (normoblastic, rubricytic) series
Additional hematologic cells Narrative describing numbers and morphology of eosinophils, basophils, mast cells, lymphocytes, plasma cells,
monocytes, and histiocytes if appropriate, with reference intervals
Stromal cells Narrative describing numbers and morphology of osteoblasts, osteoclasts, bony trabeculae, fibroblasts,
adipocytes, and endothelial cells; appearance of sinuses; presence of amyloid, granulomas, fibrosis, necrosis
Differential count Numbers of all cells and cell stages observed after performing a differential count on 300 to 1000 cells and
comparing results with reference intervals
Myeloid-to-erythroid ratio Computed from nucleated hematologic cells less lymphocytes, plasma cells, monocytes, and histiocytes
Iron stores Categorization of findings as increased, normal, or decreased iron stores
Diagnostic narrative Summary of the recorded findings and additional laboratory chemical, microbiologic, and immunoassay tests

SUMMARY
Adult hematopoietic tissue is located in the flat bones and the ends The medical laboratory scientist receives the bone marrow speci
of the long bones. Hematopoiesis occurs within the spongy tra men and prepares aspirate smears, crush preparations, imprints,
beculae of the bone adjacent to vascular sinuses. anticoagulated bone marrow smears, and fixed biopsy sections,
Bone marrow collection is a safe but invasive procedure per and specimens for confirmatory studies.
formed by a pathologist or hematologist in collaboration with a The medical laboratory scientist and pathologist collaborate with
medical laboratory scientist to obtain specimens for diagnosis residents, fellows, attending physicians, and medical laboratory
of hematologic and systemic disease and monitoring of science students to stain and review bone marrow aspirate
treatment. smears, biopsy sections, and confirmatory procedure results.
The necessity for a bone marrow examination should be evaluated Confirmatory procedures include cytochemistry, cytogenetics, and
in light of all clinical and laboratory information. In anemias for immunophenotyping by flow cytometry; fluorescence in situ
which the cause is apparent from the RBC indices, a bone marrow hybridization; and molecular diagnostics.
examination is not required. Examples of indications for bone The medical laboratory scientist and pathologist determine cellu
marrow examination include multilineage abnormalities in the larity and megakaryocyte distribution, then perform a differential
peripheral blood, pancytopenia, circulating blasts, and staging of count of 300 to 1000 bone marrow hematopoietic cells and compute
lymphomas and carcinomas. the M:E ratio, comparing the results with reference intervals.
A peripheral blood specimen is collected for a complete blood The pathologist characterizes features of hematopoietic disease,
count no more than 24 hours before the bone marrow is collected metastatic tumor cells, and abnormalities of the bone marrow
and the results of the tests are reported together. stroma and prepares a systematic written bone marrow examina
Bone marrow may be collected from the posterior or anterior iliac tion report including a diagnostic narrative.
crest or sternum using sterile disposable biopsy and aspiration
needles and cannulas. The site and equipment depend on how old Now that you have completed this chapter, go back and
the patient is and whether both an aspirate and a biopsy specimen read again the case study at the beginning and respond
are desired. to the questions presented.

R E V I E W Q UESTIONS
1. Where is most hematopoietic tissue found in adults? 2. What is the preferred bone marrow collection site in adults?
a. Lungs a. Second intercostal space on the sternum
b. Spleen b. Anterior or posterior iliac crest
c. Flat bones c. Any of the thoracic vertebrae
d. Long bones d. Anterior head of the femur
CHAPTER 16 Bone Marrow Examination 225

3. The aspirate should be examined under low power to 8. Which of the following is not an indication for a bone
assess all of the following except: marrow examination?
a. Cellularity a. Pancytopenia (reduced numbers of RBCs, WBCs, and
b. Megakaryocyte numbers platelets in the peripheral blood)
c. Morphology of abnormal cells b. Anemia with RBC indices corresponding to low serum
d. Presence of tumor cell clusters iron and low ferritin levels
c. Detection of blasts in the peripheral blood
4. What is the normal M:E ratio range in adults? d. Need for staging of Hodgkin lymphoma
a. 1.5:1 to 3.3:1
b. 5.1:1 to 6.2:1 9. In a bone marrow biopsy specimen, the RBC precursors
c. 8.6:1 to 10.2:1 were estimated to account for 40% of the cells in the
d. 10:1 to 12:1 marrow, and the other 60% were granulocyte precursors.
What is the M:E ratio?
5. What is the most common erythrocytic stage found in a. 4:6
normal marrow? b. 1.5:1
a. Pronormoblasts c. 1:1.5
b. Pronormoblasts and basophilic normoblasts d. 3:1
c. Basophilic and polychromatophilic normoblasts
d. Polychromatophilic and orthochromic normoblasts 10. On a bone marrow core biopsy sample, several large cells
with multiple nuclei were noted. They were located close
6. What cells, occasionally seen in bone marrow biopsy to the endosteum, and their nuclei were evenly spaced
specimens, are responsible for the formation of bone? throughout the cell. What are these cells?
a. Macrophages a. Megakaryocytes
b. Plasma cells b. Osteoclasts
c. Osteoblasts c. Adipocytes
d. Osteoclasts d. Fibroblasts

7. What is the largest hematopoietic cell found in a normal 11. The advantage of a core biopsy bone marrow sample over
bone marrow aspirate? an aspirate is that the core biopsy specimen:
a. Osteoblast a. Can be acquired by a less invasive collection
b. Myeloblast technique
c. Pronormoblast b. Permits assessment of the architecture and cellular
d. Megakaryocyte arrangement
c. Retains the staining qualities of basophils owing to
the use of Zenker fixative
d. Is better for the assessment of bone marrow iron
stores with Prussian blue stain

REFERENCES
1. Abboud CN, Lichtman MA: Structure of the marrow and clinical hematology, ed 11, Philadelphia, 2004, Lippincott
the hematopoietic microenvironment. In Lichtman MA, Williams & Wilkins.
Beutler E, Kipps TJ, et al, editors: Williams hematology, ed 7, 7. Ryan DH, Felgar RE: Examination of the marrow. In
New York, 2006, McGraw-Hill, pp 35-72. Lichtman MA, Beutler E, Kipps TJ, et al, editors: Williams
2. Farhi DC, Chai CC, Edelman AS, et al: Pathology of bone hematology, ed 7, New York, 2006, McGraw-Hill, pp 21-31.
marrow and blood cells, Philadelphia, 2004, Lippincott 8. Krause JR: Bone marrow biopsy, New York, 1981, Churchill
Williams & Wilkins. Livingstone.
3. Foucar K: Bone marrow pathology, ed 2, Chicago, 2001, ASCP 9. Hartsock RJ, Smith EB, Pett CS: Normal variations with
Press. aging of the amount of hemopoietic tissue in bone marrow
4. Ryan DH, Cohen HJ: Bone marrow examination. In Hoffman from the anterior iliac crest. Am J Clin Pathol 43:326-331,
R, Benz EJ, Shattil SJ, et al, editors: Hematology basic principles 1965.
and practice, Philadelphia, 2005, Churchill Livingstone, 10. Abrahamson JF, Lund-Johansen F, Laerum OD, et al: Flow
pp 2656-2671. cytometric assessment of peripheral blood contamination
5. Reddy V, Marques MB, Fritsma GA: Quick guide to hematology and proliferative activity of human bone marrow cell popula-
testing, Washington, DC, 2007, AACC Press. tions. Cytometry 58B:25, 1995.
6. Perkins SL: Examination of the blood and bone marrow.
In Greer JP, Foerster J, Lukens JN, et al, editors: Wintrobes
17 Body Fluids in the
Hematology Laboratory
Leilani Collins

OUTLINE OBJECTIVES
Performing Cell Counts After completion of this chapter, the reader will be able to:
on Body Fluids
Preparing Cytocentrifuge 1. Describe the method for performing cell counts on 6. Identify from written descriptions normal cells found
Slides body fluids. in cerebrospinal, serous, and synovial fluids.
Cerebrospinal Fluid 2. Given a description of a body fluid for cell counting, 7. Describe the characteristics of benign versus malig-
Gross Examination choose the appropriate diluting fluid, select a count- nant cells in body fluids and recognize written
Cell Counts ing area, and calculate and correct (if necessary) the descriptions of each.
Differential Cell Counts counts. 8. Differentiate exudates and transudates based on
Serous Fluid 3. Using the proper terminology, discuss the gross formation (cause), specific gravity, protein concen-
Transudates Versus appearance of body fluids, including its significance tration, appearance, and cell concentration.
Exudates and its practical use in determining cell count 9. Identify crystals in synovial fluids from written
Gross Examination
dilutions. descriptions, including polarization characteristics.
Differential Cell Counts
4. Discuss the advantages and disadvantages of cyto- 10. Describe the process of obtaining bronchoalveolar
Synovial Fluid
Gross Examination centrifuge preparations. lavage (BAL) samples, including safety precautions
Differential Cell Counts 5. Differentiate between traumatic spinal tap and for analysis; state the purpose of BAL; and recognize
Crystals cerebral hemorrhage on the basis of cell counts types of cells that normally would be found in BAL
Bronchoalveolar Lavage and appearance of uncentrifuged and centrifuged specimens.
Specimens specimens.
Procedure and
Precautions
Differential Cell Counts

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 33-year-old semiconscious woman was brought to the Most of the cells seen on the cytocentrifuge slide were
emergency department by her husband. The previous day neutrophils.
she had complained of a headache and had left work early, 1. When multiple tubes of CSF are obtained, which tube
taken some aspirin and a nap, and felt better later in the should be used for cell counts?
evening. Her husband stated that the next morning she 2. What dilution should be made to obtain a satisfactory
couldnt talk, and he brought her to the emergency depart- cytocentrifuge slide?
ment. A spinal tap was performed. The fluid that arrived in 3. What should you look for on the cytocentrifuge slide?
the laboratory was cloudy. The WBC count was 10.6 109/L. 4. What is the most likely diagnosis for this patient?

226
CHAPTER 17 Body Fluids in the Hematology Laboratory 227

T
he analysis of body fluids, including nucleated blood (or similar) to lyse the RBCs and allow an accurate count. If
cell count and differential count, can provide valuable the fluid is blood-tinged to slightly bloody, the RBCs can be
diagnostic information. This chapter is not intended as counted using undiluted fluid, but it is advisable to use a
a comprehensive treatment of all body fluids, but it covers cell small (1:2) dilution with Trk solution (or similar) to lyse the
counting and morphologic hematology. The fluids discussed RBCs and provide an accurate WBC or nucleated cell count. If
in this chapter include cerebrospinal fluid (CSF), serous or the fluid is bloody, a 1:200 dilution with isotonic saline for
body cavity fluids (pleural, pericardial, and peritoneal fluids), RBCs and either a 1:2 or a 1:20 dilution with Trk solution
and synovial (joint) fluids. Bronchoalveolar lavage (BAL) spec- for nucleated cells should be used to obtain an accurate
imens are discussed briefly. count. When performing dilutions for blood cell counts, a
calibrated pipette should be used, such as MLA pipettes or the
Ovation BioNatural pipettes (VistaLab Technologies, Inc., Mt.
PERFORMING CELL COUNTS ON
Kisco, N.Y.).
BODY FLUIDS
The number of squares to be counted on the hemacytom-
Examination of all fluids should include observation of color eter should be determined on the basis of the number of cells
and turbidity, determination of cell counts, and white blood present. In general, all nine squares on both sides of the hema-
cell (WBC) evaluation. Blood cell counts should be performed cytometer should be counted. If the number of cells is high,
and cytocentrifuge slides should be prepared as quickly as pos- however, fewer squares may be counted.5 Each square equals
sible after collection of the specimen, because WBCs begin to 1mm2. The formula for calculating the number of cells (see
deteriorate within 30 minutes after collection.1 It is important Chapter 14) is:
to mix the specimen gently but thoroughly before every mani
pulation (i.e., counting cells, preparing any dilution, and pre- Cells counted depth factor dilution factor
paring cytocentrifuge slides). Cell counts on fluids usually are Area counted (mm 2 )
performed using a hemacytometer (see Chapter 14); however,
some automated instruments now are capable of performing Guidelines for counting are summarized in Table 17-1.
blood cell counts on fluids. Automated cell counts are limited
by the poor sensitivity of automated methods for specimens
PREPARING CYTOCENTRIFUGE SLIDES
with low counts. Each instrument manufacturer should provide
a statement of intended use that defines which body fluids have The cytocentrifuge enhances the ability to identify the types of
been approved by a regulatory agency for testing on the instru- cells present in a fluid. This centrifuge spins at a low speed,
ment.2,3 Care should be taken to observe the operating limits which minimizes distortion of the cellular elements and pro-
of these instruments and the volume limits for the fluid vides a button of cells that are concentrated into a small area.
received. Red blood cell (RBC) counts on serous and synovial The cytocentrifuge assembly consists of a cytofunnel, filter
fluids have little clinical value4; relevant clinical information is paper to absorb excess fluid, and a glass slide. These three
obtained merely from the appearance of the fluid (grossly components are fastened together in a clip assembly, a few
bloody, bloody, slightly bloody). drops of well-mixed specimen are dispensed into the cytofun-
Cell counts are performed with undiluted fluid if the fluid nel, and the entire assembly is centrifuged slowly. The cells are
is clear. If the fluid is hazy or bloody, appropriate dilutions deposited onto the slide, and excess fluid is absorbed into the
should be made to permit accurate counts of WBCs and RBCs. filter paper, which produces a monolayer of cells in a small
The smallest reasonable dilution should be made. The diluting button (Figure 17-1).
fluid for RBCs is isotonic saline. Diluting fluids for WBCs Although there is some cell loss into the filter paper, this is
include glacial acetic acid to lyse the RBCs, or Trk solution, not selective, and an accurate representation of the types of
which contains glacial acetic acid and methylene blue to stain cells present in a fluid is provided. There also may be some
the nuclei of the WBCs. Acetic acid cannot be used for synovial distortion of cells as a result of the centrifugation process or
fluids, because synovial fluid contains hyaluronic acid, which crowding of cells when high cell counts are present. To mini-
coagulates in acetic acid. A small amount of hyaluronidase mize distortion resulting from overcrowding of cells, appro
powder (a pinch, or what can be picked up between two priate dilutions should be made with normal saline before
wooden sticks) or one drop of 0.05% hyaluronidase in phos- centrifugation. The basis for this dilution should be the
phate buffer per milliliter of fluid should be added to the WBC count or the nucleated cell count. A nucleated cell count
synovial fluid sample to liquefy it before performing cell counts of 200/mm3 or fewer provides a good basis for the differential.
or preparing cytocentrifuge slides. Dilutions should be based If the RBC count is extremely elevated, a larger dilution
on the turbidity of the fluid or on the number of cells seen on may be necessary; however, an RBC count of 5000/mm3 would
the hemacytometer when using an undiluted sample. not cause significant nucleated cell distortion. If a fluid has a
A WBC count of approximately 200/mm3 or an RBC count nucleated cell count of 2000/mm3 and an RBC count of
of approximately 400/mm3 causes a fluid to be slightly hazy. 10,000/mm3, a 1:10 dilution should be made, which produces
If the fluid is blood-tinged to slightly bloody, the RBCs can be a nucleated cell count of 200/mm3 and an RBC count of
counted using undiluted fluid, but for WBCs or nucleated cells 1000/mm3 for the cytocentrifuge slide. If the RBC count of a
it is advisable to use a small (1:2) dilution with Trk solution fluid is greater than 1 million/mm3, it is best to make a push
228 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 17-1 Guidelines for Counting Fluids


GROSS APPEARANCE
Test Clear Hazy Blood-Tinged Cloudy Bloody
WBCs 0-200/mm 3
>200/mm 3
Unknown High Unknown
Dilution for counting cells None 1:2 Trk 1:2 Trk 1:20 in Trk solution 1:2 in Trk solution
No. squares to count on 9 9 9 9 or 4 9 or 4
hemacytometer
RBCs 0-400/mm3 Unknown >400/mm3 Unknown >6000/mm3
Dilution for counting cells None None None None 1:200 in isotonic saline
No. squares to count on 9 large 9 large 9 or 4 large 4 large or 5 small 5 small
hemacytometer
Cytospin dilution (0.25mL Undiluted Dilute with saline to Straight or by Dilute with saline to Dilute by nucleated cell count; if RBC
[5 drops] of fluid)* 100-200/mm3 nucleated 100-200/mm3 count >1 million/mm3, make a push
nucleated cell cell count nucleated cell smear and differentiate cells that
count count are pushed out on the end

RBC, Red blood cell; WBC, white blood cell.


*Expected cell yield (WBC count for no. of cells recovered on slide): 0/mm3 for 0-70, 1-2/mm3 for 12-100, >3/mm3 for >100.

the differential count is not important, but if the number of


nucleated cells present is small, use of a systematic meander
starting at one side of the button and working toward the other
side is best. In case the number of cells recovered is small, the
area around the cell button should be marked on the back of
the slide with a wax pencil, or premarked slides should be used
to prepare cytocentrifuge slides (see Figure 17-1).

CEREBROSPINAL FLUID
CSF is the only fluid that exists in quantities sufficient to
sample in healthy individuals. CSF is present in volumes of
100 to 150mL in adults, 60 to 100mL in children, and 10 to
60mL in newborns.6,7 This fluid bathes the brain and spinal
column and serves as a cushion to protect the brain, as a cir-
culating nutrient medium, as an excretory channel for nervous
tissue metabolism, and as lubrication for the central nervous
system.
Figure 17-1 Wright-stained cytocentrifuge slide showing a concentrated
button of cells within a marked circle. (From Carr JH, Rodak BF: Clinical Gross Examination
hematology atlas, ed 3, Philadelphia, 2009, Saunders.) Normal CSF is nonviscous, clear, and colorless. A cloudy or
hazy appearance may indicate the presence of WBCs (greater
slide to perform the differential. In this case, the differential than 200/mm3), RBCs (greater than 400/mm3), or micro
should be performed on the cells pushed out on the end of organisms.6,7 Bloody fluid may be caused by a traumatic tap,
the smear instead of in the body of the smear, because that is in which blood is acquired as the puncture is performed, or by
where the larger, and possibly more significant, cells would be a pathologic hemorrhage within the central nervous system. If
deposited. more than one tube is received, the tubes can be observed for
If a consistent amount of fluid is used when cytocentrifuge clearing from tube to tube. If the first tube contains blood but
slides are prepared, a consistent yield of cells can be expected; the remaining tubes are clear or progressively clearer, the blood
this can be used to confirm the WBC or nucleated cell count. is the result of a traumatic puncture. If all tubes are uniformly
For example, if 5 drops of fluid (undiluted or diluted) is always bloody, the probable cause is a subarachnoid hemorrhage.
used to prepare cytocentrifuge slides, a 100-cell differential When a bloody sample is received, an aliquot should be
count should be obtainable if the WBC or nucleated cell count centrifuged, and the color of the supernatant should be
is equal to or greater than 3/mm3. In all cases, the entire cell observed and reported. A clear, colorless supernatant indicates
button should be scanned before the differential count is per- a traumatic tap, whereas a yellowish or pinkish yellow tinge
formed to ensure that significant clumps of cells are not over- may indicate a subarachnoid hemorrhage. This yellowish
looked. The area of the cell button that is used for performing color sometimes is referred to as xanthochromia, but because
CHAPTER 17 Body Fluids in the Hematology Laboratory 229

CSF
Gross appearance
Cloudy
Bloody
Clear
Yellow

Report
Spin and report
Supernatant appearance

Cell counts

Cloudy
Clear or bloody

Count undiluted Dilute

Trk (1:2) RBC


or (1:200)
(1:20)

Cytospin slide Report RBC


Report WBC
Figure 17-2 Flow chart for examination of cerebrospinal fluid (CSF). RBC, Red blood cell; WBC, white blood cell. (Modified from Kjeldsberg CR, Knight JA:
Body fluids, ed 3, Chicago, 1993, ASCP Press; reprinted with permission.)

TABLE 17-2 Characteristics of Cerebrospinal Fluid WBCB


RBCCSF
= WBCs added by traumatic puncture
RBCB
Traumatic Tap Pathologic Hemorrhage
Clear supernatant Colored or hemolyzed supernatant where WBCB is the WBC count for peripheral blood, RBCCSF is
Clearing from tube to tube Same appearance in all tubes the RBC count for CSF, and RBCB is the RBC count for peri
Bone marrow contamination Erythrophages pheral blood. The corrected or true CSF WBC count (WBCCSF)
Cartilage cells Siderophages (may have bilirubin crystals) is calculated as follows5:

RBC, Red blood cell. True WBCCSF


= CSF WBC hemacytometer count WBCs added

Some laboratories have questioned the value of an RBC


not all xanthochromia is pathologic, the Clinical and Labora- count on CSF and report only the WBC count.
tory Standards Institute recommends avoiding the term and A high WBC count may be found in fluid from patients with
simply reporting the actual color of the supernatant (Figure infective processes, such as meningitis. In general, WBC counts
17-2 and Table 17-2).5 are much higher (in the thousands) in patients with bacterial
meningitis than in patients with viral meningitis (in the hun-
Cell Counts dreds).8 The predominant cell type present on the cytocentri-
When multiple tubes of spinal fluid are collected, the cell count fuge slide (neutrophils or lymphocytes), however, is a better
is generally performed on tube 3, or the tube with the lowest indicator of the type of meningitisbacterial or viral. Elevated
possibility of peripheral blood contamination. Normal cell WBC or nucleated cell counts also may be obtained in patients
counts in CSF are 0 to 5WBCs/mm3 and 0RBCs/mm3. If a with inflammatory processes and malignancies.
high RBC count is obtained, one may determine whether the
source of WBCs is peripheral blood contamination by using Differential Cell Counts
the peripheral blood ratio of 1WBC per 500 to 900RBCs. If The cells normally seen in CSF are lymphocytes and monocytes
peripheral blood cell counts are known, the number of blood (Figure 17-3). In adults, the predominant cells are lympho-
WBCs added to the CSF sample can be calculated using the cytes; in newborns, the predominant cells are monocytes.6,7
following formula: Neutrophils are not normal in CSF but may be seen in small
230 PART III Routine Laboratory Evaluation of Blood Cells

Figure 17-3 Monocyte (left) and lymphocyte (right) seen in normal cere- Figure 17-5 Reactive (viral) lymphocytes in cerebrospinal fluid from a
brospinal fluid (1000). patient with viral meningitis (1000).

A B

B C

Figure 17-6 Eosinophil (A), lymphocytes (B), monocyte (C), and neutro-
Figure 17-4 Neutrophil with bacteria in cerebrospinal fluid from a patient
phil (D) in cerebrospinal fluid from a patient with a shunt (1000).
with bacterial meningitis (Wright stain, 1000). (From Carr JH, Rodak BF:
Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)

numbers because of concentration techniques. When the WBC


count is elevated and large numbers of neutrophils are seen, a
thorough and careful search should be made for bacteria,
because organisms may be present in very small numbers early
in bacterial meningitis (Figure 17-4). In viral meningitis, the
predominant cells seen are lymphocytes, including reactive or
viral lymphocytes and plasmacytoid lymphocytes (Figure
17-5). Eosinophils and basophils may be seen in response to
the presence of foreign materials such as shunts, in parasitic
infections, or in allergic reactions (Figure 17-6).6,7 When nucle-
ated RBCs are seen, bone marrow contamination resulting
from accidental puncture of the vertebral body during spinal
tap should be suspected and reported. In the case of bone
marrow contamination, other immature neutrophils and Figure 17-7 Clump of ependymal cells in cerebrospinal fluid (200).
megakaryocytes also may be seen. When there is obvious bone
marrow contamination, the WBC differential is likely to be diagnostically significant, it is important not to confuse them
equivalent to that of the bone marrow and not that of the CSF. with malignant cells (Figure 17-7).
Ependymal and choroid plexus cells, lining cells of the Cartilage cells may be seen if the vertebral body is acciden-
central nervous system, may be seen. These are large cells with tally punctured. These cells usually occur singly, are medium
abundant cytoplasm that stains lavender with Wright stain. to large, and have cytoplasm that stains wine red with a deep
They most often appear in clumps, and although they are not wine red nucleus with Wright stain (Figure 17-8).
CHAPTER 17 Body Fluids in the Hematology Laboratory 231

Figure 17-8 Cartilage cells in cerebrospinal fluid (400). Figure 17-10 Lymphoblasts in cerebrospinal fluid (1000). (From Carr JH,
Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)

Figure 17-9 Siderophage with bilirubin crystals (hematoidin) in cerebro-


spinal fluid (400). Figure 17-11 Clump of breast tumor cells in cerebrospinal fluid (400).

Siderophages are macrophages (i.e., monocytes or histio- contamination and not central nervous system involvement.
cytes) that have ingested RBCs and, as a result of the breakdown The possibility of peripheral blood contamination should be
of the RBCs, contain hemosiderin. Hemosiderin appears as large, reported and the tap should be repeated in a few days.
rough-shaped, dark blue or black granules in the cytoplasm Malignant cells resulting from metastases to the central
of the macrophage. These cells also may contain bilirubin or nervous system may be found. The most common primary
hematoidin crystals, which are golden yellow and are a result of tumors that metastasize to the central nervous system in adults
further breakdown of the ingested RBCs. The presence of sidero- are breast, lung, and gastrointestinal tract tumors and mela-
phages indicates a pathologic hemorrhage. Siderophages appear noma.6,7 In children metastases to the central nervous system
approximately 48 hours after hemorrhage and may persist for are related to Wilms tumor, Ewing sarcoma, neuroblastoma,
2 to 8 weeks after the hemorrhage has occurred (Figure 17-9). and embryonal rhabdomyosarcoma.7 Malignant cells are
A high percentage of patients with acute lymphoblastic leu- usually large with a high nucleus-to-cytoplasm ratio and are
kemia or acute myelocytic leukemia have central nervous often basophilic or hyperchromic. They often occur in clumps
system involvement.6,7 It is always important to look carefully but may occur singly. Within clumps of malignant cells, there
for leukemic cells (i.e., blast forms) in the CSF of patients with is dissimilarity between cells, and in multinucleated cells, there
leukemia. Patients with lymphoma, myeloma, and chronic may be variation in nuclear size. Clumps of malignant cells
myelogenous leukemia in blast crisis also may have blast cells may appear three-dimensional, requiring up-and-down focus-
in the CSF. These blast cells have the characteristics of blast ing to see the cells on different planes, and there are usually
forms in the peripheral blood, including a high nucleus-to- no windows between the cells. The nuclei of these cells are
cytoplasm ratio, a fine stippled nuclear chromatin pattern, usually large, often with abnormal distribution of chromatin,
and prominent nucleoli. They are usually large cells that stain and they may have an indistinct or jagged border, or there may
basophilic with Wright stain and have a fairly uniform appear- be blebbing at the border. Increased mitosis may be shown
ance (Figure 17-10). If a traumatic tap has occurred and the by the presence of several mitotic figures in the cell button.
patient has a high blast count in the peripheral blood, the Malignant cells frequently have a bizarre appearance (Figure
blasts seen in the CSF may be the result of peripheral blood 17-11 and Table 17-3).9
232 PART III Routine Laboratory Evaluation of Blood Cells

TABLE 17-3 Characteristics of Benign and Malignant TABLE 17-4 Serous Fluid: Transudates versus Exudates
Cells
Characteristic Transudates Exudates
Benign Malignant
Specific gravity <1.016 >1.016
Occasional large cells. Many cells may be very large. Protein <3g/dL >3g/dL
Light to dark staining. May be very basophilic. Lactate dehydrogenase <200IU >200IU
Rare mitotic figures. May have several mitotic figures. White blood cells <1000/mm3 >1000/mm3
Round to oval nucleus; nuclei are May have irregular or bizarre (predominant cell
uniform in size with varying nuclear shape. type mononuclear)
amounts of cytoplasm. Protein: fluid-to-serum ratio <0.5 >0.5
Nuclear edge is smooth. Edges of nucleus may be Lactate dehydrogenase: <0.6 >0.6
indistinct and irregular. fluid-to-serum ratio
Nucleus is intact. Nucleus may be disintegrated at Color Clear or straw-colored Cloudy or yellow,
edges. amber, or
Nucleoli are small, if present. Nucleoli may be large and grossly bloody
prominent. Volume Extremely large
In multinuclear cells (mesothelial), Multinuclear cells have varying
all nuclei have similar sizes and shapes of nuclei.
appearance (size and shape).
Moderate to small N:C ratio. May have high N:C ratio. Gross Examination
Clumps of cells have similar Clumps of cells contain cells of Transudates should appear straw-colored and clear. A cloudy
appearance among cells, are varying sizes and shapes, are or hazy fluid may indicate an exudate from an infectious
in the same plane of focus, three-dimensional (require process; a bloody fluid, trauma or malignancy; and a milky
and may have windows focusing up and down to fluid, effused chyle in the pleural cavity.
between cells. see all cells), and have
dark-staining borders. Differential Cell Counts
The cells found in normal serous fluid are lymphocytes, mono-
N:C, Nucleus-to-cytoplasm.
histiocytes (macrophages), and mesothelial cells. Neutrophils
commonly are seen in the fluid sent to the laboratory for
analysis but would not be present in normal fluid. When
neutrophils are seen, they have more segments and longer
SEROUS FLUID
filaments than in peripheral blood (Figure 17-13).
Serous fluids, including pleural, pericardial, and peritoneal Mesothelial cells are the lining cells of body cavities and are
fluids, normally exist in very small quantities and serve as shed into these cavities constantly. These are large (12- to
lubricant between the membranes of an organ and the sac 30-m) cells and have a fried egg appearance with basophilic
in which it is housed. Pleural fluid is found in the space cytoplasm, oval nucleus with smooth nuclear borders, stippled
between the lungs and the pleural sac; pericardial fluid, in nuclear chromatin pattern, and one to three nucleoli.6,7 Meso-
the space between the heart and the pericardial sac; and peri- thelial cells may vary in size, may be multinucleated (including
toneal fluid, between the intestine and the peritoneal sac giant cells with 20 to 25 nuclei), and may have frayed cytoplas-
(Figure 17-12). An accumulation of fluid in a cavity is termed mic borders, cytoplasmic vacuoles, or both. They may occur
an effusion. When an effusion is in the peritoneal cavity, it singly, in small or large clumps, or in sheets. When they occur
also may be referred to as ascites or ascitic fluid.4 It would be in clumps, there are usually windows between the cells. The
difficult to remove these fluids from a healthy individual; nucleus-to-cytoplasm ratio is 1:2 to 1:3, and this is generally
the presence of these fluids in detectable amounts indicates a consistent despite the variability in cell size.6 They tend to have
disease state. a similar appearance to each other on a slide. Mesothelial cells
are seen in most effusions, and their numbers are increased in
Transudates Versus Exudates sterile inflammations and decreased in tuberculous pleurisy
As noted, the accumulation of a large amount of fluid in a and bacterial infections (Figure 17-14).6
cavity is called an effusion. Effusions are subdivided further into Macrophages appear as monocytes or histiocytes in serous
transudates and exudates to distinguish whether disease is fluids and may contain RBCs (erythrophages) or siderotic
present within or outside the body cavity. In general, transu- granules (siderophages), or they may appear as signet ring
dates develop as part of systemic disease processes, such as cells when lipid has been ingested and the resulting large
congestive heart failure, whereas exudates indicate disorders vacuole pushes the nucleus to the periphery of the cell
associated with bacterial or viral infections, malignancy, pul- (Figure 17-15).
monary embolism, or systemic lupus erythematosus. Several Eosinophils and basophils are not normally seen. These
parameters can be measured to determine whether an effusion may be present in large numbers, however, as a result of aller-
is a transudate or an exudate (Table 17-4). gic reaction or sensitivity to foreign material.
CHAPTER 17 Body Fluids in the Hematology Laboratory 233

Lung
Parietal membrane
Visceral membrane (pericardium)
(pleura)
Pericardial cavity

Pleural cavity
Visceral membrane
Parietal membrane (pericardium)
(pleura)

Body wall
Parietal membrane
(peritoneum) Liver

Peritoneal cavity Stomach

Visceral membrane
(peritoneum) Intestine

Figure 17-12 Parietal and visceral membranes of the pleural, pericardial, and peritoneal cavities. Parietal membranes line the body wall, whereas visceral
membranes enclose organs. The two membranes are actually one continuous membrane. The space between opposing surfaces is identified as the body
cavity (i.e., pleural, pericardial, and peritoneal cavities). (From Brunzel NA: Fundamentals of urine and body fluid analysis, ed 2, Philadelphia, 2004,
Saunders.)

TABLE 17-5 Gram-Stained Organisms Most Commonly


Seen in Body Fluids
Fluid Organism
Cerebrospinal Gram-negative diplococci
Gram-positive cocci
Gram-negative coccobacilli
Yeaststains gram-positive
Cryptococcuslook for capsule
Serous (peritoneal, pleural, Gram-positive cocci
or pericardial) Gram-negative bacilli
Gram-positive bacilli
Yeaststains gram-positive
Synovial (joint) Gram-positive cocci
Figure 17-13 Hypersegmented neutrophil with prominent filaments. Gram-negative bacilli
Normal appearance of neutrophils in body fluids (1000). (From Carr JH, Gram-negative diplococci
Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
Gram-negative coccobacilli

NOTE: If the Gram-stained organisms seen in a fluid are not listed above for that fluid,
When large numbers of neutrophils are seen, a thorough do not report Gram stain results. Save the slide for review.
search should be made for bacteria. If possible, Gram staining
should be performed on a second cytocentrifuge slide to aid in factors necessary for the formation of these cellspresence of
rapid identification if bacteria are found. Table 17-5 lists Gram- the lupus erythematosus factor, incubation, and trauma to the
stained organisms most commonly seen in body fluids. cellsexist in vivo. A lupus erythematosus cell is an intact neu-
Lupus erythematosus cells may be seen in serous fluids of trophil that has engulfed a homogeneous mass of degenerated
patients with systemic lupus erythematosus, because all the nuclear material, which displaces the normal nucleus. Lupus
234 PART III Routine Laboratory Evaluation of Blood Cells

A A

B B
Figure 17-14 A, Mesothelial cells in peritoneal fluid (200). Note Figure 17-15 A, Erythrophage in peritoneal fluid (1000). B, Signet ring
fried egg appearance. B, Mesothelial cell with 21 nuclei in pleural fluid cell (arrow) in peritoneal fluid (200).
(400).

erythematosus cells can form in vivo and in vitro in serous


and synovial fluids and should be reported (Figure 17-16).
Malignant cells are seen in serous fluids from primary or
metastatic tumors. They have the characteristics of malignant
cells found in CSF (Figure 17-17). Figure 17-18 presents a flow
chart for examination of serous fluids.

SYNOVIAL FLUID
Gross Examination
Synovial fluid is normally present in very small amounts in the
synovial cavity surrounding joints. When fluid is present in
amounts large enough to aspirate, there is a disease process in
the joint. Normally this fluid is straw-colored and clear. Syno-
Figure 17-16 Lupus erythematosus cell (arrow) in pleural fluid (1000).
vial fluid contains hyaluronic acid, which makes it very viscous.
A small amount of hyaluronidase powder should be added to
all joint fluids to liquefy them before cell counts are performed
or cytocentrifuge slides are prepared. If a crystal analysis is to the synovial cavity and are shed into the cavity. They resemble
be performed, an aliquot of fluid should be removed for this mesothelial cells but are usually present in smaller numbers
purpose before the hyaluronidase is added. (Figure 17-19).
Lupus erythematosus cells may be present in synovial fluid
Differential Cell Counts just as in serous fluid. Malignant cells are rarely seen in syno-
Cells found in normal synovial fluid are lymphocytes, vial fluid, but when present resemble tumor cells seen in serous
monocytes/histiocytes, and synovial cells. Synovial cells line fluids or CSF.
A B

C D
Figure 17-17 A, Clump of tumor cells in pleural fluid (200). B, Tumor cells and mitotic figure in pleural fluid (1000). C, Adenocarcinoma cells in pleural
fluid (200). D, Tumor cells in peritoneal fluid (200). Note cannibalism.

Serous fluids
(Pleural, peritoneal, pericardial, ascitic)

Gross appearance (color, clarity)

Cell counts

WBC RBC

Trk (1:2) or Undiluted or


(1:20) (1:200)
dilution with saline

Cytospin slide

If dilution is necessary, base dilution on NCC. Dilute to 100-200/mm3

Normal Segmented leukocytes Tumor

Lymphocyte Look for bacteria Send to cytology laboratory


Histiocytes/monocytes
Mesothelial cells
Perform gram stain
on cytocentrifuge slide

Report results
Figure 17-18 Flow chart for examination of serous fluid. NCC, Nucleated cell count; RBC, red blood cell; WBC, white blood cell.
236 PART III Routine Laboratory Evaluation of Blood Cells

A
Figure 17-19 Synovial cells in synovial fluid (400). Note similarity to
mesothelial cells.

B
Figure 17-21 Intracellular (A) and extracellular (B) monosodium urate
crystals in synovial fluid (1000). A, Wright stain. B, Polarized with red
A compensator. (A from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
Philadelphia, 2009, Saunders. B courtesy George Girgis, MT[ASCP], Indiana
University Health, Indianapolis, Ind.)

a cytocentrifuge preparation. However, the specimen should


be fresh, without hyaluronidase added. All synovial fluids
should be examined carefully for crystals using a polarizing
microscope with a red compensator. The crystals most com-
monly seen in synovial fluids are cholesterol, calcium pyro-
phosphate, and monosodium urate.
Cholesterol crystals are large, flat, extracellular crystals with
a notched corner.10 They are seen in patients with chronic effu-
sions, particularly patients with rheumatoid arthritis.
Calcium pyrophosphate crystals are seen in pseudogout.
B
These crystals are intracellular and are small rhomboid,
Figure 17-20 Intracellular calcium pyrophosphate crystals in synovial platelike, or rodlike crystals.10 The crystals are weakly birefrin-
fluid (1000). A, Wright stain. B, Polarized with red compensator. (B courtesy gent when polarized (i.e., they do not appear bright when
George Girgis, MT[ASCP], Indiana University Health, Indianapolis, Ind.) polarized). When the red compensator is used, calcium pyro-
phosphate crystals appear blue when the longitudinal axis of
Many neutrophils are present in synovial fluid in acute the crystal is parallel to the y-axis (Figure 17-20).10
inflammation of joints. As always, a careful search should be Monosodium urate crystals are seen in gout. They are large
made for bacteria when many neutrophils are seen. needlelike crystals that may be intracellular or extracellular.
These crystals are strongly birefringent when polarized. When
Crystals the red compensator is used, monosodium urate crystals
Intracellular and extracellular crystals may be present in syno- appear yellow when the longitudinal axis of the crystal is paral-
vial fluid. Crystal examination may be performed by placing a lel to the y-axis (Figure 17-21).10 Figure 17-22 presents a flow
drop of fluid on a slide and adding a coverslip or by examining chart for synovial fluid analysis.
CHAPTER 17 Body Fluids in the Hematology Laboratory 237

Synovial (joint) fluids

Gross appearance (color, clarity)

Add hyaluronidase Crystals


(polarize to confirm)

1. Monosodium urate
Cell counts (needlelike)
RBC 2. Calcium pyrophosphate
(rhomboid)
WBC 3. Cholesterol
Undiluted or (notched wedge)
(1:200)
Trk (1:2) or dilution with saline
(1:20)

Cytospin slide

If dilution is necessary, base dilution on NCC. Dilute to 100-200/mm3

Normal Segmented leukocytes

Lymphocytes Look for bacteria Look for LE cells


Histiocytes/monocytes
Synovial cells
Perform Gram stain Report LE
on cytocentrifuge slide cells seen on
cytocentrifuge slide

Report results
Figure 17-22 Flow chart for examination of synovial (joint) fluid. LE, Lupus erythematosus; NCC, nucleated cell count; RBC, red blood cell; WBC, white
blood cell.

obtained, some hematology laboratories no longer perform


BRONCHOALVEOLAR LAVAGE SPECIMENS
this procedure and defer to information reported from the
Procedure and Precautions microbiology laboratory.
BAL specimens are not naturally occurring fluids; they are Cell counts and cytocentrifuge preparations are performed
produced when the BAL procedure is performed. The proce- as with any body fluid. Significant cell deterioration occurs
dure consists of introducing warmed saline into the lungs within 30 minutes of collection, with the neutrophils disinte-
in 50-mL aliquots and then withdrawing it. The specimen grating most rapidly.
received in the laboratory is the withdrawn fluid. The purpose
of the procedure is to determine types of organisms and cells
that are present in areas of the lung that are otherwise inacces- Differential Cell Counts
sible. This procedure is performed on patients with severe The cell types most commonly seen in BAL specimens are
lung dysfunction. The specimen should always undergo an neutrophils, monohistiocytes (macrophages), and lympho-
extensive microbiologic workup and often cytologic examina- cytes. Mesothelial cells are not seen in BAL specimens, because
tion. It is common to see bacteria, yeast, or both on cytocen- these cells line the body cavities and not the interior of the
trifuge slides prepared from these specimens. Because samples lung. Pneumocytes, which can resemble mesothelial cells or
are obtained from the interior of the lung and may contain adenocarcinoma, may be seen in patients with adult respira-
airborne organisms, care should be taken to avoid aerosol tory distress syndrome.
production. Samples should be mixed and containers opened Ciliated epithelial cells can be seen and should be reported,
under a biologic safety hood, and a mask should be worn because they indicate that the sample was obtained from the
when performing cell counts. Because the risk of performing upper respiratory tract instead of deeper in the lung. These are
cell counts and preparing cytocentrifuge slides on BAL speci- columnar cells with the nucleus at one end of the cell, elon-
mens outweighs the clinical relevance of the information gated cytoplasm, and cilia at the opposite end of the cell from
238 PART III Routine Laboratory Evaluation of Blood Cells

Figure 17-23 Ciliated epithelial cells in bronchoalveolar lavage fluid Figure 17-24 Histiocytes with carbonaceous material in bronchoalveolar
(100). lavage fluid (40).

the nucleus. They can occur in clusters. If the sample is not


aged when the cell count is performed, these cells are in motion
in the hemacytometer, because they can be propelled by their
cilia (Figure 17-23).
Histiocytes laden with carbonaceous material are seen
in patients who smoke tobacco. These cells resemble side
rophages in other fluids, but the carbonaceous material
is black, brown, or blue-black and is more dropletlike
(Figure 17-24).
Pneumocystis jiroveci (formerly Pneumocystis carinii) may be
seen in specimens from patients infected with human immu-
nodeficiency virus. The P. jiroveci organisms appear as clumps
of amorphous material. Close examination of the clumps may Figure 17-25 Cyst of Pneumocystis jiroveci (formerly Pneumocystis
reveal cysts (Figure 17-25). carinii) in bronchoalveolar lavage fluid (100).

SUMMARY
Cell counts and differential counts performed on body fluid speci- Bacteria and yeast may be seen in any fluid.
mens are valuable diagnostic tools. Malignant cells may be seen in any fluid but are rare in synovial
Calibrated methods must be used when performing cell counts to fluid.
provide accurate counts. Synovial fluid should be examined for crystals using a polarizing
To optimize cell morphologic features, specimens should not be microscope.
overdiluted or underdiluted when cytocentrifuge slides are BAL specimens are not a true body fluid, but examination of cells
prepared. present may provide diagnostic information.
Normal cell types in any fluid are lymphocytes, macrophages
(monocytes, histiocytes), and lining cells (ependymal cells in Now that you have completed this chapter, go back and
CSF, mesothelial cells in serous fluids, synovial cells in joint read again the case study at the beginning and respond
fluids). to the questions presented.

R E V I E W Q UESTIONS
Refer to the following scenario to answer questions 1 and 2: A squares counted on both sides. Undiluted fluid is used to
spinal fluid specimen is diluted 1:2 with Trk solution to perform the RBC count. A total of 105RBCs is counted on both
perform the nucleated cell count. Six nucleated cells are sides of the hemacytometer, with four large squares on both
counted on both sides of the hemacytometer, with all nine sides counted.
CHAPTER 17 Body Fluids in the Hematology Laboratory 239

1. The nucleated cell count is ___/mm3. a. Mesothelial cells


a. 3 b. Metastatic tumor cells
b. 7 c. Cartilage cells
c. 13 d. Pneumocytes
d. 66
7. A serous fluid with a clear appearance, specific gravity of
2. The RBC count is ___/mm3. 1.010, protein concentration of 1.5g/dL, and fewer than
a. 131 500 mononuclear cells/mm3 would be considered:
b. 263 a. Infectious
c. 1050 b. An exudate
d. 5830 c. A transudate
d. Sterile
3. Based on the cell counts, the appearance of the fluid is:
a. Turbid 8. On the cytocentrifuge slide prepared from a peritoneal
b. Hemolyzed fluid sample, many large cells are seen, singly and in
c. Clear clumps. The cells have a fried egg appearance and baso-
d. Cloudy philic cytoplasm, and some are multinucleated. These cells
should be reported as:
4. All of the following cells are normally seen in CSF, serous a. Suspicious for malignancy
fluids, and synovial fluids except: b. Macrophages
a. Lining cells c. Large lymphocytes
b. Neutrophils d. Mesothelial cells
c. Lymphocytes
d. Monocytes/histiocytes (macrophages) Refer to the following scenario to answer questions 9 and 10: A
56-year-old man came to the physicians office with complaints
5. Spinal fluid was obtained from a 56-year-old woman. On of pain and swelling in his left great toe. Fluid aspirated from
receipt in the laboratory, the fluid was noted to be slightly the toe was straw-colored and cloudy. The WBC count was
bloody. When a portion of the fluid was centrifuged, the 2543/mm3. The differential consisted mainly of neutrophils
supernatant was clear. The cell counts were 5200RBCs/ and monocytes/histiocytes. Intracellular and extracellular
mm3 and 24WBCs/mm3. On the cytocentrifuge prepara- crystals were seen on the cytocentrifuge slide. The crystals
tion, several nucleated RBCs were seen. The differential were needle-shaped and, when polarized with the use of the
was 52% lymphocytes, 20% neutrophils, 22% monocytes, red compensator, appeared yellow on the y-axis.
4% myelocytes, and 2% blasts. What is the most likely
explanation for these results? 9. The crystals are:
a. Bone marrow contamination a. Cholesterol
b. Bacterial meningitis b. Hyaluronidase
c. Peripheral blood contamination c. Monosodium urate
d. Leukemic infiltration in the central nervous system d. Calcium pyrophosphate

6. A 34-year-old woman with a history of breast cancer devel- 10. This patients painful toe was caused by:
oped a pleural effusion. The fluid obtained was bloody and a. Gout
had a nucleated cell count of 284/mm3. On the cytocentri- b. Infection
fuge preparation, there were several neutrophils and a few c. Inflammation
monocytes/histiocytes. There were also several clusters of d. Pseudogout
large, dark-staining cells. These cell clumps appeared three-
dimensional and contained some mitotic figures. What
is the most likely identification of the cells in clusters?

REFERENCES
1. Glasser L: Cells in cerebrospinal fluid. Diagn Med 4:33-50, 4. Brunzel N: Fundamentals of urine and body fluid analysis, ed 2,
1981. Philadelphia, 2004, Saunders.
2. Barnes PW, Eby CS, Shiner AG: An evaluation of the utility 5. Clinical and Laboratory Standards Institute (CLSI): Body
of performing body fluid counts on the Coulter LH 750. Lab fluid analysis for cellular composition: proposed guideline.
Hematol 10:127-131, 2004. CLSI document H56-A, vol 26, no. 26. Wayne, Pa, 2006,
3. Brown W, Keeney M, Chin-Yee I, et al: Validation of body CLSI.
fluid analysis on the Coulter LH 750. Lab Hematol 9:155-159, 6. Kjeldsberg C, Knight J: Body fluids, ed 3, Chicago, 1993, ASCP
2003. Press.
240 PART III Routine Laboratory Evaluation of Blood Cells

7. Galagan K, Blomberg D, Cornbleet, PJ, et al: Color atlas 9. Cornbleet J: Microscopy of CSF and body fluids. Workshop
of body fluids, Northfield, Ill, 2006, College of American material presented at the National Meeting of the American
Pathologists. Society of Clinical Pathologists, 1991.
8. Krueger R: Meningitis: a case study. Lab Med 18:677-681, 10. Strasinger SK, DiLorenzo MS: Urinalysis and body fluids, ed 5,
1987. Philadelphia, 2008, FA Davis.
PART IV Hematopathology: Erythrocyte Disorders

Anemias: Red Blood Cell Morphology


and Approach to Diagnosis
18
Rakesh P. Mehta*

OUTLINE OBJECTIVES
Definition of Anemia After completion of this chapter, the reader will be able to:
Clinical Findings
Physiologic Adaptations 1. Define anemia and recognize laboratory results 8. Recognize the importance of reviewing the peri
Mechanisms of Anemia consistent with anemia. pheral blood film when assessing anemias and
Ineffective and Insufficient 2. Discuss the importance of the history and the distinguish the important findings.
Erythropoiesis physical examination in the diagnosis of anemia. 9. Describe the use of the red blood cell distribution
Acute Blood Loss and 3. Describe clinical signs and symptoms of anemia and width (RDW) in the diagnosis of anemias.
Hemolysis recognize them in clinical scenarios. 10. Briefly explain how the body adapts to anemia over
Laboratory Diagnosis of 4. List procedures that are commonly performed for the time and the impact on the patients experience of
Anemia detection and diagnosis of anemia. the anemia.
Complete Blood Cell 5. Distinguish among effective, ineffective, and 11. Use an algorithm incorporating the reticulocyte
Count with Red Blood
insufficient erythropoiesis when given examples. count, MCV, and RDW to narrow the differential
Cell Indices
6. Discuss the importance of the reticulocyte count in diagnosis of anemia.
Reticulocyte Count
Peripheral Blood Film the evaluation of anemia. 12. Classify given examples of variations in red blood cell
Examination 7. Characterize the three groups of anemias involving morphology as inclusions, shape changes, volume
Bone Marrow Examination decreased production that are categorized on the changes, or color changes.
Other Laboratory Tests basis of mean cell volume (MCV) and give one
Approach to Evaluating example of each.
Anemias
Reticulocyte Count and
Anemia Classification
Mean Cell Volume and
Anemia Classification CASE STUDY
Red Blood Cell After studying the material in this chapter, the reader should be able to respond to the following
Distribution Width
case study:
Hemolytic Anemias
Pathophysiologic A 45-year-old female phoned her physician and complained of fatigue, shortness of breath on
Classification exertion, and general malaise. She requested some B12 shots to make her feel better. The physician
asked the patient to schedule an appointment so that she could determine the cause of the symp-
toms before offering treatment. The hematocrit performed in the office was 20%. The physician
then requested additional laboratory tests, including a CBC with a peripheral blood film examina-
tion and a reticulocyte count.
1. Why did the physician want the patient to come to the office before she prescribed therapy?
2. How do the MCV and reticulocyte count help determine the classification of the anemia?
3. Why is the examination of the peripheral blood film important in the work-up of an
anemia?

*The foundation for this chapter is the work of Ann Bell. The author would like to express his gratitude for the opportunity to continue to amend
her fine endeavor.

241
242 PART IV Hematopathology: Erythrocyte Disorders

R
ed blood cells (RBCs) perform the vital physiologic Obtaining a good history requires carefully questioning the
function of oxygen delivery to the tissues. The hemo- patient, particularly with regard to diet, drug ingestion, expo-
globin within the erythrocyte has the remarkable sure to chemicals, occupation, hobbies, travel, bleeding history,
capacity to bind oxygen in the lungs and then release it appro- ethnic group, family history of disease, neurologic symptoms,
priately in the tissues.1 The term anemia is derived from the previous medication, jaundice, and various underlying diseases
Greek word anaimia, meaning without blood.2 A decrease in that produce anemia.4,7-9 Although inquiry in these areas can
the number of RBCs, or the amount of hemoglobin in the reveal common conditions that can lead to anemia, there are
RBCs, results in decreased oxygen delivery and subsequent numerous other possibilities as well. Therefore, a thorough
tissue hypoxia. Anemia is a commonly encountered condition discussion is required to elicit any potential cause of the
affecting an estimated 1.62 billion people worldwide.3 Anemia anemia. For example, iron deficiency can lead to an interesting
should not be thought of as a disease, but rather as a manifesta- symptom called pica.10 Patients with pica have cravings for
tion of other underlying disease processes.4,5 Therefore, the unusual substances such as ice (pagophagia), cornstarch, or
cause of all anemias should be thoroughly investigated. This clay. Alternatively, individuals with anemia may be asymptom-
chapter provides an overview of the diagnosis, mechanisms, atic, as can be seen in mild or slowly progressive anemias.
and classification of anemia. In the following chapters, each Certain features should be evaluated closely during the
anemia is discussed in detail. physical examination to provide clues to hematologic disor-
ders, such as skin (for pallor, jaundice, petechiae), eyes (for
hemorrhage), and mouth (for mucosal bleeding). The exami-
DEFINITION OF ANEMIA
nation should also look for sternal tenderness, lymphadenopa-
A functional definition of anemia is a decrease in the oxygen- thy, cardiac murmurs, splenomegaly, and hepatomegaly.4,7-9
carrying capacity of the blood. It can arise if there is insufficient Jaundice is important for the assessment of anemia, because it
hemoglobin or the hemoglobin is nonfunctional. The former may be due to increased RBC destruction, which suggests a
is the more frequent cause. hemolytic component to the anemia. Measuring vital signs is
Anemia is defined operationally as a reduction, from the also a crucial component of the physical evaluation. Patients
baseline value, in the total number of RBCs, amount of circu- experiencing a rapid fall in hemoglobin concentration typically
lating hemoglobin, and RBC mass for a particular patient. In have tachycardia (fast heart rate), whereas if the anemia is
practice, this definition is not applicable, because a patients long-standing, the heart rate may be normal due to the bodys
baseline value is rarely known.5,6 A more conventional defini- ability to compensate for the anemia.
tion is a decrease in RBCs, hemoglobin, and hematocrit below Moderate anemias (hemoglobin concentration of 7 to 10g/
the reference range for healthy individuals of the same age, sex, dL) may not produce clinical signs or symptoms if the onset
and race, under similar environmental conditions.4-8 Problems of anemia is slow.4 Depending on the patients age and cardio-
with this conventional definition may occur for several reasons. vascular state, however, moderate anemias may be associated
The reference ranges are derived from large pools of normal with pallor of conjunctivae and nail beds, dyspnea, vertigo,
individuals; however, the definition of normal is different for headache, muscle weakness, lethargy, and other symptoms.4,7-9
each of these data sets. This has led to the development of dif- Severe anemias (hemoglobin concentration of less than 7g/
ferent reference ranges, depending on which pool of individu- dL) usually produce tachycardia, hypotension, and other
als was used. Furthermore, these pools of individuals lack the symptoms of volume loss, in addition to the symptoms listed
heterogeneity required to be universally applied to all the dif- earlier. The severity of the anemia is gauged by the degree of
ferent populations.6 reduction in RBC mass, cardiopulmonary adaptation, and the
Examples of hematologic reference ranges for the adult and rapidity of progression of the anemia.4
pediatric populations are included on the inside cover of this
text. They are listed according to age and sex, but race, envi-
PHYSIOLOGIC ADAPTATIONS
ronmental, and laboratory factors can also influence the values.
Each laboratory must determine its own reference ranges based Reduced delivery of oxygen to tissues caused by reduced hemo-
on its particular instrumentation, the methods used, and the globin causes an increase in erythropoietin secretion by the
demographics and environment of its patient population. For kidneys. Erythropoietin stimulates the RBC precursors in the
the purpose of the discussion in this chapter, a patient is con- bone marrow, which leads to the release of more RBCs into
sidered anemic if the hemoglobin value falls below those listed the circulation (see Chapter 8). With persistent anemia, the
in these tables. body implements physiologic adaptations to increase the
oxygen-carrying capacity of a reduced amount of hemoglobin.
Heart rate, respiratory rate, and cardiac output are increased
CLINICAL FINDINGS
for a more rapid delivery of oxygenated blood to tissues.
The history and physical examination are important compo- In addition, the tissue hypoxia triggers an increase in RBC
nents in making a clinical diagnosis of anemia. The classic 2,3-bisphosphoglycerate that shifts the oxygen dissociation
symptoms associated with anemia are fatigue and shortness curve to the right (decreased oxygen affinity of hemoglobin)
of breath. If oxygen delivery is decreased, then patients will and results in increased delivery of oxygen to tissues (see
not have enough energy to perform their daily functions. Chapter 10).11 This is a significant mechanism in chronic
CHAPTER 18 Anemias: Red Blood Cell Morphology and Approach to Diagnosis 243

anemias that enables patients with low levels of hemoglobin shortened RBC life span. (Note that chronic blood loss leads
to remain relatively asymptomatic. With persistent and severe to iron deficiency and is covered under insufficient erythro
anemia, however, the strain on the heart can ultimately lead poiesis in the previous section.) With acute blood loss and
to cardiac failure. excessive hemolysis, the bone marrow is able to increase pro-
duction of RBCs, but the level of response is inadequate to
compensate for the excessive RBC loss. Numerous causes of
MECHANISMS OF ANEMIA
hemolysis exist, including intrinsic defects in the RBC mem-
The life span of the RBC in the circulation is about 120 days. brane, enzyme systems, or hemoglobin, or extrinsic causes
In a healthy individual with no anemia, each day approxi- such as antibody-mediated processes, mechanical fragmenta-
mately 1% of the RBCs are removed from circulation due to tion, or infection-related destruction.4,8,12
senescence, but the bone marrow continuously produces RBCs
to replace those lost. Hematopoietic stem cells develop into
LABORATORY DIAGNOSIS OF ANEMIA
erythroid precursor cells, and the bone marrow appropriately
releases reticulocytes that mature into RBCs in the peripheral Complete Blood Cell Count with Red Blood
circulation. Adequate RBC production requires several nutri- Cell Indices
tional factors, such as iron, vitamin B12, and folate. Globin To detect the presence of anemia, the medical laboratory pro-
synthesis also must function normally. In conditions with fessional performs a complete blood count (CBC) using an
excessive bleeding or hemolysis, the bone marrow must automated hematology analyzer to determine the RBC count,
increase RBC production to compensate for the increased RBC hemoglobin concentration, hematocrit, RBC indices, white
loss. Therefore, the maintenance of a stable hemoglobin blood cell (WBC) count, and platelet count. The RBC indices
concentration requires the production of functionally normal include the mean cell volume (MCV), mean cell hemoglobin
RBCs in sufficient numbers to replace the amount lost.4,7,8 (MCH), and mean cell hemoglobin concentration (MCHC)
(see Chapter 14).13 The most important of these indices is the
Ineffective and Insufficient Erythropoiesis MCV, a measure of the average RBC volume in femtoliters (fL).
Erythropoiesis is the term used for marrow erythroid prolifera- Reference ranges for these determinations are listed on the
tive activity. Normal erythropoiesis occurs in the bone marrow inside front cover of the text. Automated hematology analyzers
(see Chapter 8).4 When erythropoiesis is effective, the bone also provide an RBC histogram and the red blood cell distribu-
marrow is able to produce functional RBCs that leave the tion width (RDW). A relative and absolute reticulocyte count,
marrow and supply the peripheral circulation with adequate described subsequently, should be performed for every patient
numbers of cells. when anemia is found. Automated analyzers are available to
Ineffective erythropoiesis refers to the production of erythroid perform reticulocyte counts with greater accuracy and precision
progenitor cells that are defective. These defective progenitors than manual counting methods.
are often destroyed in the bone marrow before their matu The RBC histogram is an RBC volume frequency distribu-
ration and release into the peripheral circulation. Several tion curve with the relative number of cells plotted on the
conditions, such as megaloblastic anemia, thalassemia, and ordinate and RBC volume in femtoliters on the abscissa. With
sideroblastic anemia, are characterized by ineffective erythro- a normal population of RBCs, the distribution is approxi-
poiesis. In these anemias, the peripheral blood hemoglobin is mately gaussian. Abnormalities include a shift in the curve to
low despite an increase in RBC precursors in the bone marrow. the left (microcytosis) or to the right (macrocytosis), and a
The effective production rate is considerably less than the total widening of the curve caused by a greater variation of RBC
production rate, which results in a decreased number of normal volume about the mean or by the presence of two populations
circulating RBCs. Consequently, the patient becomes anemic.12 of RBCs with different volumes (anisocytosis). The histogram
Insufficient erythropoiesis refers to a decrease in the number of complements the peripheral blood film examination in iden-
erythroid precursors in the bone marrow, resulting in decreased tifying variant RBC populations.12 (A discussion of histograms
RBC production and anemia. Several factors can lead to the with examples can be found in Chapter 39.)
decreased RBC production, including a deficiency of iron (inad- The RDW is the coefficient of variation of RBC volume
equate intake, malabsorption, excessive loss from chronic expressed as a percentage.13 It indicates the variation in RBC
bleeding); a deficiency of erythropoietin, the hormone that volume within the population measured and correlates with
stimulates erythroid precursor proliferation and maturation anisocytosis on the peripheral blood film. Automated analyz-
(renal disease); loss of the erythroid precursors due to an auto- ers calculate the RDW by dividing the standard deviation
immune reaction (aplastic anemia, acquired pure red cell of the RBC volume by the MCV and then multiplying by 100
aplasia) or infection (parvovirus B19); or suppression of the to convert to a percentage. The usefulness of the RDW is
erythroid precursors due to infiltration of the bone marrow with discussed later.
granulomas (sarcoidosis) or malignant cells (acute leukemia).12
Reticulocyte Count
Acute Blood Loss and Hemolysis The reticulocyte count serves as an important tool to assess the
Anemia can also develop as a result of acute blood loss (such bone marrows ability to increase RBC production in response
as traumatic injury) or premature hemolysis resulting in a to an anemia. Reticulocytes are young RBCs that lack a nucleus
244 PART IV Hematopathology: Erythrocyte Disorders

TABLE 18-1 Formulas for Reticulocyte Counts and Red Blood Cell Indices
Test Formula Adult Reference Range
9
Absolute reticulocyte count ( 10 /L) = [reticulocytes (%)/100] RBC count ( 10 /L)
12
25-75 109/L
Corrected reticulocyte count (%) = reticulocytes (%) patients Hct (%)/45
Reticulocyte production index (RPI) = corrected reticulocyte count/maturation time In anemic patients, RPI should be >3
Mean cell volume (MCV) (fL) = Hct (%) 10/RBC count ( 1012/L) 80-100fL
Mean cell hemoglobin (MCH) (pg) = Hb (g/dL) 10/RBC count ( 1012/L) 26-32pg
Mean cell hemoglobin concentration (MCHC) (g/dL) = Hb (g/dL) 100/Hct (%) 32-36g/dL

Hb, Hemoglobin; Hct, hematocrit; RBC, red blood cell.

but still contain residual ribonucleic acid (RNA). Normally, production of normal RBCs, due to either insufficient or inef-
they circulate peripherally for only 1 day while completing fective erythropoiesis. Reticulocytes and related calculations
their development. The adult reference range for the reticulo- are discussed in Chapter 14.
cyte count is 0.5% to 1.5% expressed as a percentage of the
total number of RBCs.13 The newborn reference range is 1.5% Peripheral Blood Film Examination
to 5.8%, but these values change to approximately those of an An important component in the evaluation of an anemia is
adult within a few weeks after birth.7-9 An absolute reticulocyte examination of the peripheral blood film, with particular atten-
count is determined by multiplying the percent reticulocytes tion to RBC diameter, shape, color, and inclusions. The periph-
by the RBC count. The reference range for the absolute reticu- eral blood film also serves as a quality control to verify the
locyte count is 25 to 75 109/L, based on a normal adult RBC results produced by automated analyzers. Normal RBCs on a
count.4 A patient with a severe anemia may seem to be produc- Wright-stained blood film are nearly uniform, ranging from 6
ing increased numbers of reticulocytes if only the percentage to 8m in diameter. Small or microcytic cells are less than
is considered. For example, an adult patient with 1.5 1012/L 6m in diameter, and large or macrocytic RBCs are greater
RBCs and 3% reticulocytes has an absolute reticulocyte count than 8m in diameter. Certain shape abnormalities of diag-
of 45 109/L. The percentage of reticulocytes is above the refer- nostic value (such as sickle cells, spherocytes, schistocytes, and
ence range, but the absolute reticulocyte count is within the oval macrocytes) and RBC inclusions (such as malarial para-
reference range. For the degree of anemia, however, both of sites, basophilic stippling, and Howell-Jolly bodies) can be
these results are inappropriately low. In other words, produc- detected only by studying the RBCs on a peripheral blood film
tion of reticulocytes within the reference range is inadequate (Tables 18-2 and 18-3). Examples of abnormal shapes and
to compensate for an RBC count that is approximately one inclusions are provided in Figure 18-1.
third of normal. Finally, a review of the WBCs and platelets may help show
The reticulocyte count may be corrected for anemia by that a more generalized bone marrow problem is leading to
multiplying the reticulocyte percentage by the patients hema- the anemia. For example, hypersegmented neutrophils can be
tocrit and dividing the result by 45 (the average normal seen in vitamin B12 or folate deficiency, whereas blast cells and
hematocrit). If the reticulocytes are released prematurely from decreased platelets may be an indication of acute leukemia.
the bone marrow and remain in the circulation 2 to 3 days (See Chapter 15 for a complete discussion of the peripheral
(instead of 1 day), the corrected reticulocyte count must be blood film evaluation.) Additional information from the blood
divided by maturation time to determine the reticulocyte pro- film examination always complements the data from the auto-
duction index (RPI). The RPI is a better indication of the rate mated hematology analyzer.
of RBC production than is the corrected reticulocyte count
(Table 18-1).4 Bone Marrow Examination
Analysis of the reticulocyte count plays a crucial role in The cause of many anemias can be determined from the history,
determining whether an anemia is due to an RBC production physical examination, and results of laboratory tests on periph-
defect or to a premature destruction and shortened survival eral blood. When the cause cannot be determined, however, or
defect. If there is shortened RBC survival, as in the hemolytic the differential diagnosis remains broad, a bone marrow aspira-
anemias, the bone marrow tries to compensate by increasing tion and biopsy may help in establishing the cause of anemia.4,8
RBC production. This increased production of RBCs results in A bone marrow examination is indicated for a patient with an
the release of more reticulocytes into the peripheral circulation unexplained anemia associated with or without other cytope-
and a higher reticulocyte count. Although an increased reticu- nias, fever of unknown origin, or suspected hematologic malig-
locyte count can also be observed in acute blood loss, it is more nancy. A bone marrow examination evaluates hematopoiesis
commonly observed in the hemolytic anemias.4,13 Chronic and can determine if there is abnormal infiltration of the
blood loss, on the other hand, does not lead to an appropriate marrow. Important findings in the marrow that can point to
increase in the reticulocyte count, but rather leads to iron the underlying cause of the anemia include abnormal cellular-
deficiency and a subsequent low reticulocyte count. An inap- ity of the marrow (e.g., hypocellularity in aplastic anemia);
propriately low reticulocyte count results from decreased evidence of ineffective erythropoiesis and megaloblastic
CHAPTER 18 Anemias: Red Blood Cell Morphology and Approach to Diagnosis 245

TABLE 18-2 Description of Red Blood Cell (RBC) Abnormalities and Commonly Associated Disease States
RBC Abnormality Cell Description Commonly Associated Disease States
Anisocytosis Abnormal variation in RBC volume or diameter Hemolytic, megaloblastic, iron deficiency anemia
Macrocyte Large RBC (>8m in diameter), MCV >100fL Megaloblastic anemia
Myelodysplastic syndrome
Chronic liver disease
Bone marrow failure
Reticulocytosis
Oval macrocyte Large oval RBC Megaloblastic anemia
Microcyte Small RBC (<6m in diameter), MCV <80fL Iron deficiency anemia
Anemia of chronic inflammation
Sideroblastic anemia
Thalassemia
HbE disease and trait
Poikilocytosis Abnormal variation in RBC shape Severe anemia
Certain shapes helpful diagnostically
Spherocyte Small, round, dense RBC with no central pallor Hereditary spherocytosis
Immune hemolytic anemia
Extensive burns (along with schistocytes)
Elliptocyte, ovalocyte Elliptical (cigar-shaped), oval (egg-shaped), RBC Hereditary elliptocytosis or ovalocytosis
Iron deficiency anemia
Thalassemia major
Myelophthisic anemias
Stomatocyte RBC with slitlike area of central pallor Hereditary stomatocytosis
Rh deficiency syndrome
Acquired stomatocytosis (liver disease, alcoholism)
Artifact
Sickle cell Thin, dense, elongated RBC pointed at each end; may be Sickle cell anemia
curved Sickle cell-thalassemia
Hb C crystal Hexagonal crystal of dense hemoglobin formed within the Hb C disease
RBC membrane
Hb SC crystal Fingerlike or quartzlike crystal of dense hemoglobin Hb SC disease
protruding from the RBC membrane
Target cell (codocyte) RBC with hemoglobin concentrated in the center and around Liver disease
the periphery resembling a target Hemoglobinopathies
Thalassemia
Schistocyte (schizocyte) Fragmented RBC due to rupture in the peripheral circulation Microangiopathic hemolytic anemia* (along with
microspherocytes)
Traumatic cardiac hemolysis
Extensive burns (along with microspherocytes)
Helmet cell (keratocyte) RBC fragment in shape of a helmet Same as schistocyte
Folded cell RBC with membrane folded over Hb C disease
Hb SC disease
Acanthocyte (spur cell) Small, dense RBC with few irregularly spaced projections of Severe liver disease (spur cell anemia)
varying length Neuroacanthocytosis (abetalipoproteinemia,
McLeod syndrome)
Burr cell (echinocyte) RBC with blunt or pointed, short projections that are usually Uremia
evenly spaced over the surface of cell; present in all fields Pyruvate kinase deficiency
of blood film but in variable numbers per field
Teardrop cell (dacryocyte) RBC with a single pointed extension resembling a teardrop Primary myelofibrosis
or pear Myelophthisic anemia
Thalassemia
Megaloblastic anemia

Hb, Hemoglobin; MCV, mean cell volume.


*Such as thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, disseminated intravascular coagulation.

Cells with similar morphology that are unevenly distributed in a blood film (not present in all fields) are likely due to a drying artifact in blood film preparation; these artifacts are
sometimes called crenated RBCs.
246 PART IV Hematopathology: Erythrocyte Disorders

TABLE 18-3 Erythrocyte Inclusions: Description, Composition, and Some Commonly Associated Disease States
Appearance in Appearance in Associated
Inclusion Supravital Stain Wright Stain Inclusion Composed of Diseases/Conditions
Diffuse basophilia Granules and filaments Bluish tinge throughout RNA Hemolytic anemia
cytoplasm; also called After treatment for iron, vitamin
polychromasia B12, or folate deficiency
Basophilic stippling Granules and filaments Blue-purple granules Precipitated RNA Lead poisoning
(punctate distributed throughout Thalassemia
basophilia) cytoplasm Hemoglobinopathies
Abnormal heme synthesis
Howell-Jolly body Dense, round, granule Dense, round, blue or purple DNA (nuclear fragment) Hyposplenism
granule; usually one per After splenectomy
cell; occasionally multiple Megaloblastic anemia
Hemolytic anemia
Heinz body Round granule attached Not visible Denatured hemoglobin Glucose-6-phosphate
to inner membrane dehydrogenase deficiency
Unstable hemoglobins
Oxidant drugs/chemicals
Pappenheimer bodies* Clusters of small granules Clusters of small, light blue Iron Sideroblastic anemia
granules, often near Hemoglobinopathies
periphery of cell Hyposplenism
Megaloblastic anemia
Cabot ring Rings or figure-eights Blue rings or figure-eights Remnant of mitotic spindle Megaloblastic anemia
Myelodysplastic syndromes
Hb H Fine, evenly dispersed Not visible Precipitate of chains of Hb H disease
granules hemoglobin

Hb, Hemoglobin.
*Blue (siderotic) granules observed in Prussian blue stain.

changes (e.g., vitamin B12 deficiency or myelodysplastic syn- reticulocyte count and a microcytic anemia are present. Serum
drome); lack of iron on iron stains of the bone marrow (the vitamin B12 and serum folate assays are helpful in investigating
gold standard for diagnosis of iron deficiency); and the pres- a macrocytic anemia with a low reticulocyte count, whereas a
ence of granuloma, fibrosis, infectious agents, and tumor cells direct antiglobulin test can differentiate autoimmune hemo-
that may be inhibiting normal erythropoiesis. lytic anemias from hemolytic anemias with other causes.
Other tests that can assist in the diagnosis of anemia can be Because of the numerous potential etiologies of anemia, the
performed on the bone marrow sample as well, including flow cause needs to be determined before initiation of therapy.7,12
cytometry, cytogenetic studies, and molecular analysis to detect
abnormal cells, specific gene mutations, and chromosome
APPROACH TO EVALUATING ANEMIAS
abnormalities. (Chapter 16 discusses bone marrow procedures
and bone marrow examination in detail.) The approach to the patient with anemia begins with taking a
complete history and performing a physical examination.4,5,7-9
Other Laboratory Tests For example, new-onset fatigue and shortness of breath suggest
Other laboratory tests that can assist in establishing the cause an acute drop in the hemoglobin concentration, whereas a
of anemia include routine urinalysis (to detect hemoglobinuria lack of symptoms suggests a long-standing or congenital condi-
or an increase in urobilinogen) with a microscopic examina- tion. A strict vegetarian may not be getting enough vitamin
tion (to detect hematuria or hemosiderin) and analysis of stool B12 in the diet, whereas an individual with alcoholism may
(to detect occult blood or intestinal parasites). Also, certain not be getting enough folate. A large spleen may be an indica-
chemistry studies are very useful, such as renal and hepatic tion of hereditary spherocytosis, whereas a stool positive
function tests. More recently, copper deficiency has been iden- for occult blood may indicate iron deficiency. The complete
tified as another nutritional cause of anemia.14 history and physical examination can yield information to
After the hematologic laboratory studies are completed, the narrow the possible cause or causes of the anemia and thus
anemia may be classified based on reticulocyte count, MCV, lead to a more rational approach to ordering the appropriate
and peripheral blood film findings. Iron studies (including diagnostic tests.
serum iron, total iron-binding capacity, transferrin saturation, The first step in the laboratory diagnosis of anemia is
and serum ferritin) are valuable if an inappropriately low detecting its presence by the accurate measurement of the
Normal Spherocytes Burr cells

Target cells Ovalocytes Elliptocytes

Sickle cells Hb C crystals Hb SC crystals

Acanthocytes Schistocytes Teardrop cells

Stomatocytes Howell-Jolly bodies Basophilic stippling


Figure 18-1 Red blood cells (RBCs): varied RBC shapes and inclusions. Hb, Hemoglobin. (Modified from Carr JH, Rodak BF: Clinical hematology atlas,
ed 3, Philadelphia, 2009, Saunders.)
248 PART IV Hematopathology: Erythrocyte Disorders

hemoglobin, hematocrit, and RBC count and comparison of Mean Cell Volume and Anemia Classification
these values with the reference range for healthy individuals of Microcytic anemia is characterized by an MCV of less than 80fL
the same age, sex, race, and environment. Knowledge of previ- with small RBCs (less than 6m in diameter). Hypochromia,
ous hematologic results is often extremely valuable. A reduc- characterized by an MCHC of less than 32g/dL and increased
tion of 10% or more in hematologic values may be the first central pallor in the RBCs, may accompany the microcytosis.
clue that an abnormal condition may be present.4,6,15 Microcytic anemias are caused by conditions that result in
There are numerous causes of anemia, so a rational algo- reduced hemoglobin synthesis: iron deficiency, inability to
rithm to evaluate this condition is required. Several of the utilize iron (chronic inflammatory states), globin synthesis
tests already discussed help guide the approach to evaluating defect (thalassemia), and heme synthesis defect (sideroblastic
patients with anemia, including the CBC, reticulocyte count, anemia, lead poisoning). The most common microcytic anemia
RBC indices (particularly the MCV), and examination of the results from an iron level that is insufficient for maintaining
peripheral blood film. normal erythropoiesis; it is characterized by abnormal results
on iron studies. Early iron deficiency is manifested only by
Reticulocyte Count and Anemia Classification reduced iron stores without anemia or microcytosis. The causes
The absolute reticulocyte count is useful in initially classifying of iron deficiency vary in infants, children, adolescents, and
anemias into the categories of decreased RBC production adults, and it is imperative to find the cause before beginning
(decreased reticulocyte count) and shortened RBC survival treatment (see Chapter 19).
(increased reticulocyte count). When the reticulocyte count is Macrocytic anemia is characterized by an MCV greater than
decreased, the MCV can further classify the anemia into three 100fL with large RBCs (greater than 8m in diameter). Mac-
subgroups: (1) normocytic anemias, (2) microcytic anemias, rocytic anemias may be megaloblastic or nonmegaloblastic.
and (3) macrocytic anemias. Figure 18-2 presents an algorithm Megaloblastic anemias are caused by conditions that impair
that illustrates how anemias can be evaluated based on the synthesis of deoxyribonucleic acid (DNA), such as vitamin B12
reticulocyte count and MCV. and folate deficiency or myelodysplasia. Nuclear maturation

Anemia

Absolute reticulocyte count

Low or normal High

Decreased or ineffective RBC production Excessive RBC loss

MCV

Acute hemorrhage Hemolysis


Low Normal High
(Microcytic) (Normocytic) (Macrocytic) Intrinsic causes
Membrane defects
Iron deficiency Aplastic anemia Vitamin B12 Hemoglobinopathies
Anemia of chronic Anemia of renal deficiency Hb E disease/trait
inflammation disease Folate deficiency Enzyme deficiencies
Sideroblastic Infection Aplastic anemia Extrinsic causes
anemia (parvovirus B19) Immune hemolytic
Myelodysplastic anemias
Thalassemia Myelophthisic syndrome Microangiopathic hemolytic
anemia Erythroleukemia anemias (TTP, HUS, DIC)
Anemia of chronic Chronic liver Macroangiopathic hemolytic
inflammation disease anemias (traumatic cardiac hemolysis)
Infectious agents (malaria, babesiosis)
Some drugs Drugs, chemicals, venoms, extensive burns

Figure 18-2 Algorithm for evaluating causes of anemia based on absolute reticulocyte count and mean cell volume (MCV). The list of anemias contains
examples; there are numerous other causes not listed. Anemia of chronic liver disease is multifactorial and can be normocytic. DIC, Disseminated intravascular
coagulation; Hb, hemoglobin; HUS, hemolytic uremic syndrome; RBC, red blood cell; TTP, thrombotic thrombocytopenic purpura.
CHAPTER 18 Anemias: Red Blood Cell Morphology and Approach to Diagnosis 249
lags behind cytoplasmic development as a result of impaired film must be examined to rule out a dimorphic population of
DNA synthesis. This asynchrony between nuclear and cytoplas- microcytes and macrocytes that can yield a normal MCV. The
mic development results in larger cells. All cells of the body are presence of a dimorphic population can also be verified by
ultimately affected by the defective production of DNA (see observing a bimodal distribution on the RBC histogram pro-
Chapter 20). Pernicious anemia is one cause of vitamin B12 duced by an automated CBC instrument (see Chapter 39).
deficiency, whereas malabsorption secondary to inflammatory Some normocytic anemias develop due to the premature
bowel disease is one cause of folate deficiency. A megaloblastic destruction and shortened survival of RBCs (hemolytic
anemia is characterized by oval macrocytes and hyperseg- anemias), and they are characterized by an elevated reticulo-
mented neutrophils in the peripheral blood and by megalo- cyte count. Other normocytic anemias develop due to a
blasts or large nucleated RBC precursors in the bone marrow. decreased production of RBCs and are characterized by a
The MCV in megaloblastic anemia can be markedly increased decreased reticulocyte count. Figure 18-3 presents an algorithm
(up to 150fL), but modest increases (100 to 115fL) occur for initial evaluation of anemia based on the MCV.
as well.
Nonmegaloblastic forms of anemia are also characterized Red Blood Cell Distribution Width
by large RBCs, but in contrast to megaloblastic anemias, they The RDW can help determine the cause of an anemia when
are typically related to membrane changes owing to disruption used in conjunction with the MCV. Each of the three MCV
of the cholesterol-to-phospholipid ratio. These macrocytic cells categories mentioned previously (normocytic, microcytic, mac-
are mostly round, and the marrow nucleated RBCs do not rocytic) can also be subclassified by the RDW as homogeneous
display the megaloblastic maturation changes. Macrocytic (normal RDW) or heterogeneous (increased or high RDW),
anemias are often seen in patients with chronic liver disease according to Bessman etal.16,17 For example, a low MCV with
and bone marrow failure. It is rare for the MCV to be greater a high RDW is suggestive of iron deficiency (Table 18-4). This
than 115fL in these nonmegaloblastic anemias. classification is not absolute, however, because there can be an
Normocytic anemia is characterized by an MCV in the range overlap of RDW values among some of the conditions in each
of 80 to 100fL. The RBC morphology on the peripheral blood MCV category.

Classification of Anemia Based on


Red Blood Cell Mean Volume (MCV)

Microcytic Macrocytic Normocytic


MCV  80 fL MCV  100 fL MCV 80-100 fL

Sideroblastic Iron Anemia of Thalassemia Retic Retic N or


anemia deficiency chronic Hb E disease/trait
anemia inflammation

Nonmegaloblastic Megaloblastic

Aplastic anemia Hemolytic anemia Aplastic anemia


Chronic liver disease Anemia of renal disease
Alcoholism Myelophthisic anemia
Infection (parvovirus B19)
Anemia of chronic inflammation

Vitamin B12 Folate Myelodysplasia Intrinsic Extrinsic


deficiency deficiency Erythroleukemia
Some drugs

Membrane Hemoglobinopathies Enzyme Immune Nonimmune RBC injury:


defects deficiencies causes Microangiopathic
Macroangiopathic
Infectious agents
Other injury: drugs,
chemicals, venoms,
extensive burns
Figure 18-3 Algorithm for classification of anemia based on mean cell volume (MCV). Anemia of chronic liver disease is multifactorial and can be normo
cytic. , Increased; , decreased; Hb, hemoglobin, N, normal; retic, absolute reticulocyte count.
250 PART IV Hematopathology: Erythrocyte Disorders

TABLE 18-4 Classification of Anemia Based on Red Blood Cell Mean Volume (MCV) and Red Blood Cell
Distribution Width (RDW)
MCV LOW MCV NORMAL MCV HIGH
RDW Normal RDW High RDW Normal RDW High RDW Normal RDW High
Heterozygous Iron deficiency Anemia of chronic Early iron, folate, or Aplastic anemia Folate or vitamin B12
thalassemia Sickle cell- inflammation vitamin B12 deficiency Chronic liver disease deficiency
Anemia of chronic thalassemia Anemia of renal disease Mixed deficiency of iron Alcoholism Myelodysplastic syndrome
inflammation Acute hemorrhage + vitamin B12 or folate Chemotherapy
Hb E disease/trait Hereditary spherocytosis Sickle cell anemia Cold agglutinin disease
Hb SC disease Chronic liver disease

Modified from Bessman JD, Gilmer PR, Gardner FH: Improved classification of anemias by MCV and RDW, Am J Clin Pathol 80:324, 1983; and Marks PW, Glader B: Approach
to anemia in the adult and child. In Hoffman R, Benz EJ, Shattil SJ, etal, editors. Hematology: basic principles and practice, ed 5, Philadelphia, 2009, Churchill Livingstone.
Hb, Hemoglobin; RDW, red blood cell distribution width.
When the RDW is normal, the red blood cells (RBCs) are homogeneous in volume; when the RDW is high, the RBCs are heterogeneous in volume.

BOX 18-1 Pathophysiologic Classification of Anemias

Anemia Caused by Decreased Production of Red Blood Cells


Hematopoietic stem cell failure: acquired and hereditary aplastic anemia
Functional impairment of erythroid progenitors:
Disturbance of DNA synthesis: megaloblastic anemia
Disturbance of hemoglobin synthesis: iron deficiency anemia, thalassemia, sideroblastic anemia, anemia of chronic inflammation
Disturbance of proliferation and differentiation of erythroid precursors: anemia of renal failure, anemia associated with marrow infiltration

Anemia Caused by Increased Red Blood Cell Destruction or Loss


Intrinsic abnormality
Membrane defect: hereditary spherocytosis, hereditary elliptocytosis, pyropoikilocytosis, paroxysmal nocturnal hemoglobinuria
Enzyme deficiency: glucose-6-phosphate dehydrogenase deficiency, pyruvate kinase deficiency
Globin abnormality: sickle cell anemia, other hemoglobinopathies
Extrinsic abnormality
Immune causes: warm-type autoimmune hemolytic anemia, cold agglutinin disease, paroxysmal cold hemoglobinuria, transfusion reaction,
hemolytic disease of the fetus and newborn
Nonimmune red blood cell injury: microangiopathic hemolytic anemia (thrombotic thrombocytopenic purpura, hemolytic uremic syndrome,
HELLP syndrome, disseminated intravascular coagulation), macroangiopathic hemolytic anemia (traumatic cardiac hemolysis), infectious agents
(malaria, babesiosis, bartonellosis, clostridial sepsis), other injury (chemicals, drugs, venoms, extensive burns)
Blood loss: acute blood loss anemia

HELLP, Hemolysis, elevated liver enzymes, and low platelets syndrome.


The list of anemias is not all-inclusive; there are numerous other conditions not listed.
Modified from Prchal JT: Clinical manifestations and classification of erythrocyte disorders. In Lichtman MA, Beutler E, Kipps TJ, etal, editors: Williams hematology,
ed 7, New York, 2006, McGraw-Hill. Available at: http://www.accessmedicine.com/content.aspx?aID=2139970. Accessed March 1, 2010.

Hemolytic Anemias Pathophysiologic Classification


In cases of an elevated reticulocyte count caused by shortened In a pathophysiologic classification of anemia, related condi-
survival of RBCs, an investigation for a hemolytic anemia is tions are grouped by the mechanism causing the anemia. In
required. As indicated previously, numerous intrinsic and this classification scheme, the anemias caused by decreased
extrinsic causes of hemolysis exist. A direct antiglobulin test RBC production (e.g., disorders of DNA synthesis) are distin-
helps differentiate immune-mediated destruction from the guished from the anemias caused by increased RBC destruction
other causes. In the other hemolytic anemias, reviewing the or loss (intrinsic and extrinsic abnormalities of RBCs). Box
peripheral blood film is vital for determining the cause of 18-1 presents a pathophysiologic classification of anemia
the hemolysis (see Table 18-2 and Figure 18-1). Hemolytic based on the causes of the abnormality and gives one or more
anemias are discussed in Chapters 22 to 26. examples of an anemia in each classification.
CHAPTER 18 Anemias: Red Blood Cell Morphology and Approach to Diagnosis 251

SUMMARY
Anemia is defined conventionally as a decrease in RBCs, hemo- and bone marrow examination, if indicated. Other tests are indi-
globin, and hematocrit below the reference range for healthy cated based on the RBC indices, history, and physical examination
individuals of the same age, sex, and race, under similar environ- (e.g., serum iron, total iron-binding capacity, serum ferritin, serum
mental conditions. folate and vitamin B12, and direct antiglobulin test).
Clinical diagnosis of anemia is based on history, physical examina- The reticulocyte count and MCV play crucial roles in investigation
tion, signs, symptoms, and laboratory test results. of the cause of an anemia.
Many anemias have common manifestations. Careful questioning Subclassifications of anemias based on MCV include normocytic,
of the patient may reveal contributing factors, such as diet, medi- microcytic, and macrocytic anemias.
cations, occupational hazards, and bleeding history. The MCV, when combined with the RDW, also can aid in diagnos-
A thorough physical examination is valuable in determining the ing anemia.
cause of anemia. Some of the areas that should be evaluated are The peripheral blood film plays an important role in the diagnosis
skin, nail beds, eyes, mucosa, lymph nodes, heart, and size of the of hemolytic anemias.
spleen and liver. Anemias may have more than one pathophysiologic cause.
Moderate anemias may not manifest clinical symptoms if the The cause of anemia should be determined before treatment is
onset is slow. Severe anemias (hemoglobin concentration of less initiated.
than 7g/dL) usually produce pallor, dyspnea, vertigo, headache,
muscle weakness, lethargy, hypotension, and tachycardia. Now that you have completed this chapter, go back and
Laboratory procedures helpful in the diagnosis of anemia include read again the case study at the beginning and respond
CBC with RBC indices and RDW, reticulocyte count, examination to the questions presented.
of the peripheral blood film with emphasis on RBC morphology,

R E V I E W Q UESTIONS
1. Common clinical signs and symptoms of anemia include: 5. An increase in which one of the following suggests a
a. Chills and fever reduced RBC life span and a hemolytic anemia?
b. Jaundice and enlarged lymph nodes a. Reticulocyte count
c. Shortness of breath and fatigue b. RDW
d. Splenomegaly c. Hemoglobin
d. Hematocrit
2. The RBC index that is used to describe the average RBC
volume is the: 6. An increased MCV, along with a high RDW, suggests:
a. RDW a. Iron deficiency anemia
b. MCV b. Vitamin B12 or folate deficiency
c. MCH c. Sickle cell anemia
d. MCHC d. Normal blood picture

3. Variation in RBC volume is expressed by the: 7. Which of the following is detectable only by examination
a. RDW of a peripheral blood film?
b. MCV a. Microcytosis
c. MCH b. Anisocytosis
d. MCHC c. Poikilocytosis
d. Hypochromia
4. An anemia develops because the bone marrow becomes
fibrotic and the amount of active bone marrow, including 8. Refer to Figure 18-2 and Table 18-4. Which of the follow-
RBC precursors, is diminished. The cells that are present are ing would be within the differential diagnosis of a patient
normal in appearance, but there are too few to meet the with an MCV of 115fL and an RDW of 20% (reference
demand for blood cells, and anemia develops. The reticu- range 11.5% to 14.5%)?
locyte count is low. The anemia would be described as: a. Aplastic anemia
a. Effective erythropoiesis b. Sickle cell anemia
b. Ineffective erythropoiesis c. Iron deficiency
c. Insufficient erythropoiesis d. Folate deficiency
252 PART IV Hematopathology: Erythrocyte Disorders

9. When anemia is long-standing, which of the following is 10. Which of the following patients would be considered
among the adaptations of the body? anemic with a hemoglobin value of 14.5g/dL? Refer to
a. Reduced respiratory rate reference ranges inside the front cover of this text.
b. Reduced oxygen affinity of hemoglobin a. A newborn boy
c. Lower heart rate b. An adult woman
d. Reduction in the volume of blood ejected from the c. An adult man
heart with each contraction d. A 10-year-old girl

REFERENCES
1. Schecther AN: Hemoglobin research and the origins of 10. Kettaneh A, Eclache V, Fain O, et al: Pica and food craving in
molecular medicine. Blood 112:3927-3938, 2008. patients with iron-deficiency anemia: a case-control study in
2. The American heritage dictionary of the English language, ed 4, France. Am J Med 118:185-188, 2005.
Boston, 2001, Houghton Mifflin. 11. Hsia CCW: Respiratory functions of hemoglobin. N Engl J
3. de Benoist B, McLean E, Egli I, et al: Worldwide prevalence Med 338:239-247, 1998.
of anaemia 1993-2005: WHO global database on anaemia, 12. Adamson JW, Longo DL: Anemia and polycythemia. In Fauci
Geneva, 2008, WHO Press. Available at: http://whqlibdoc. AS, Braunwald E, Kasper DL, et al, editors: Harrisons principles
who.int/publications/2008/9789241596657_eng.pdf. of internal medicine, ed 17, New York, 2008, McGraw-Hill,
Accessed July 30, 2009. pp 355-363.
4. Means RT Jr, Glader B: Anemia: general considerations. In 13. Perkins SL: Examination of the blood and bone marrow. In
Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes
clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
Health/Lippincott Williams & Wilkins, pp 779-809. Health/Lippincott Williams & Wilkins, pp 1-20.
5. Tefferi A: Anemia in adults: a contemporary approach to 14. Halfdanarson TR, Kumarm N, Li CY, et al: Hematologic
diagnosis. Mayo Clin Proc 78:1274-1280, 2003. manifestations of copper deficiency: a retrospective review.
6. Beutler E, Waalen J: The definition of anemia: what is the Eur J Haemotol 80:523-531, 2008.
lower limit of normal of the blood hemoglobin concentra- 15. Brill JR, Baumgardner DJ: Normocytic anemia. Am Fam Physi-
tion? Blood 107:1747-1750, 2006. cian 62:2255-2263, 2000.
7. Rose MG, Berliner N: Disorders of red blood cells. In 16. Bessman JD, Gilmer PR, Gardner FH: Improved classification
Andreoli TE, Carpenter CCJ, Griggs RC, Benjamin IJ, editors: of anemia by MCV and RDW. Am J Clin Pathol 80:322-326,
Cecil essentials of medicine, ed 7, Philadelphia, 2007, Saunders, 1983.
pp 496-508. 17. Marks PW, Glader B: Approach to anemia in the adult and
8. Zuckerman KS: Approach to anemias. In Arend WP, Armitage child. In Hoffman R, Benz EJ, Shattil SJ, et al, editors: Hema-
JO, Clemmons DR, et al, editors: Cecil medicine, ed 23, tology: basic principles and practice, ed 5, Philadelphia, 2009,
Philadelphia, 2007, Saunders, pp 1179-1187. Churchill Livingstone.
9. Irwin JJ, Kirchner JT: Anemia in children. Am Fam Physician
64:1379-1386, 2001.
Disorders of Iron and
Heme Metabolism
19
Kathryn Doig

OUTLINE OBJECTIVES
General Concepts in After completion of this chapter, the reader should be able to:
Anemia
Iron Deficiency Anemia 1. Recognize complete blood count (CBC) results con- 5. Recognize a bone marrow description consistent
Etiology sistent with iron deficiency anemia, anemia of with iron deficiency anemia, anemia of chronic
Pathogenesis chronic inflammation, and sideroblastic anemias. inflammation, or a sideroblastic anemia.
Epidemiology 2. Given the results of iron studies, including free eryth- 6. Recognize conditions in which anemia of chronic
Laboratory Diagnosis rocyte protoporphyrin (FEP), serum transferrin recep- inflammation may develop.
Treatment and Its Effects tor, and reticulocyte hemoglobin, distinguish findings 7. Recognize predisposing factors for sideroblastic
Anemia of Chronic consistent with iron deficiency anemia, anemia of anemias or conditions in which sideroblastic anemias
Inflammation chronic inflammation, sideroblastic anemias, thalas- may develop.
Etiology semias, and iron overload conditions. 8. Discuss the clinical significance of increased levels
Laboratory Diagnosis
3. Recognize individuals at risk for iron deficiency of FEP.
Treatment
anemia by virtue of age, gender, diet, physiologic 9. Describe the pathogenesis of iron deficiency anemia,
Sideroblastic Anemias
Lead Poisoning circumstance such as pregnancy and menstruation, anemia of chronic inflammation, sideroblastic anemia
Porphyrias or pathologic conditions such as gastric ulcers. secondary to lead poisoning, and hemochromatosis.
Iron Overload 4. Given a description of the appearance of bone marrow 10. Discuss the differences in disease etiology, diagno-
Etiology stained with Prussian blue stain, recognize results sis, and treatment between iron overload resulting
Pathogenesis consistent with iron deficiency anemia, anemia of from hereditary hemochromatosis and transfusion-
Laboratory Diagnosis chronic inflammation, or a sideroblastic anemia. related hemosiderosis.
Treatment

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

An 85-year-old slender, frail white woman was hospitalized Patient Value Reference Range
for diagnosis and treatment of anemia suspected during a MCH (pg) 18.1 25.4-34.6
routine examination by her physician. The physician noted MCHC (g/dL) 27.3 31-37
that she appeared pale and inquired about fatigue and tired- RDW (%) 20 11.5-14.5
ness. Although the patient generally felt well, she admitted Platelets ( 109/L) 165.0 150-400
to feeling slightly tired when climbing stairs. A hematocrit WBC differential Unremarkable
performed in the physicians office showed a dangerously low RBC morphology Marked anisocytosis,
value of 12%, so the patient was hospitalized for further marked poikilocytosis,
evaluation. Her CBC results are as follows: marked hypochromia,
marked microcytosis
Patient Value Reference Range
9
WBCs ( 10 /L) 8.5 4.5-11 1. What conditions should you consider based on the results
RBCs ( 1012/L) 1.66 4.3-5.9 of the CBC?
Hb (g/dL) 3.0 13.9-16.3 2. Assuming that this patient has not been diagnosed with
Hct (%) 11 39-55 anemia at any other time during her life, are any of the
MCV (fL) 66.3 80-100 Continued

253
254 PART IV Hematopathology: Erythrocyte Disorders

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

conditions listed in the answer to question 1 more likely, mobility associated with aging, are any of the conditions
based on the patients age or sex? listed in the answer to question 1 more likely than the others?
3. Assuming that the patient is otherwise healthy and experi- 4. What additional testing would you recommend? What
encing only the common declines of sight, hearing, and results do you expect for this patient?

desquamated skin and sloughed intestinal epithelium.1 Because


GENERAL CONCEPTS IN ANEMIA
the body tenaciously conserves all other iron from senescent
Anemia may result whenever red blood cell (RBC) production cells, including RBCs, daily replacement of 1mg of iron from
is impaired, RBC life span is shortened, or there is frank loss the diet maintains iron balance and supplies the bodys need
of cells. The anemias associated with iron and heme typically for RBC production as long as there is no other source of loss.
are categorized as anemias of impaired production resulting When the iron in the diet is consistently inadequate, over time
from the lack of raw materials for hemoglobin assembly. the bodys stores of iron become depleted. Ultimately, RBC
Depending on the cause, lack of available iron results in iron production slows as a result of the inability to produce hemo-
deficiency anemia or the anemia of chronic inflammation. globin. With approximately 1% of cells dying naturally each
Inadequate production of protoporphyrin leads to diminished day, the anemia becomes apparent when the production rate
production of heme and thus hemoglobin, but with a relative is insufficient for replacement of lost cells.
excess of iron. The result is sideroblastic anemia. These causes
are discussed in this chapter. Inadequate globin production Increased Need
results in the thalassemias, which are discussed separately in Iron deficiency can also develop when the level of iron intake
Chapter 27. is inadequate to meet the needs of an expanding erythron.
As discussed more extensively in Chapter 11, iron is This is the case in periods of rapid growth, such as infancy
absorbed from the diet in the small intestine, carried by trans- (especially in prematurity), childhood, and adolescence. For
ferrin to a cell in need, and brought into the cell, where it is example, although both infants and adult men need about
held as ferritin until incorporated into its final functional 1mg/day of iron, that corresponds to a much higher amount
molecule. That functional molecule may be a heme-based cyto- per kilogram of body weight for the infant. Pregnancy and
chrome, muscle myoglobin, or, in the case of developing RBCs, nursing place similar demands on the mothers body to provide
hemoglobin. Iron may be unavailable for incorporation into iron for the developing fetus or nursing infant in addition to
heme because of inadequate stores of body iron or impaired her own iron needs. In each of these instances, what had previ-
mobilization. The anemia associated with inadequate stores is ously been an adequate intake of iron for the individual
termed iron deficiency anemia, whereas the anemia resulting becomes inadequate as the need for iron increases.
from impaired iron mobilization is known as anemia of chronic
inflammation because of its association with chronic inflamma- Impaired Absorption
tory conditions, such as rheumatoid arthritis. When the iron Even when the diet is adequate in iron, the inability to absorb
supply is adequate and mobilization is unimpaired but an that iron through the enterocyte into the blood over time will
intrinsic RBC defect prevents production of protoporphyrin or result in a deficiency of iron in the body. The impairments may
incorporation of iron into it, the resulting anemia is termed be pathologic, as with malabsorption due to celiac disease. In
sideroblastic, which refers to the presence of nonheme iron in addition, diseases that decrease stomach acidity impair iron
the developing RBCs. absorption by decreasing the capacity to reduce dietary ferric
iron to the absorbable ferrous form. Some loss of acidity
accompanies normal aging. Gastrectomy or bariatric surgeries
IRON DEFICIENCY ANEMIA
can impair iron absorption. Medications such as antacids can
Etiology inhibit absorption, and others may even bind the iron in the
Iron deficiency anemia develops when the intake of iron is intestine, preventing its absorption.
inadequate to meet a standard level of demand, when the need
for iron expands, when there is impaired absorption, or when Chronic Blood Loss
there is chronic loss of hemoglobin from the body. A fourth way iron deficiency develops is with chronic hemor-
rhage or hemolysis that results in the loss of small amounts of
Inadequate Intake heme iron from the body over a prolonged period of time.
Iron deficiency anemia can develop when the erythron is Eventually anemia develops when the iron loss continually
slowly starved for iron. Each day, approximately 1mg of iron exceeds iron intake and the storage iron is exhausted. Excessive
is lost from the body, mainly in the mitochondria of heme iron can be lost through chronic gastrointestinal
CHAPTER 19 Disorders of Iron and Heme Metabolism 255

Iron Depletion

Normal Iron Status Stage 1 Stage 2 Stage 3

Storage Iron Transport Iron Functional Iron


Depletion Depletion Depletion
(Iron Deficiency Anemia)

Iron Storage Compartment

Iron Transport Compartment

Functional Iron Compartment

Laboratory test values


Hemoglobin N N N

Serum iron N N

TIBC N N

Ferritin N

Figure 19-1 Development of iron deficiency anemia. , Increased; , decreased; N, normal; TIBC, total iron-binding capacity. (Adapted from Suominen
P, Punnonen K, Rajamki A, etal: Serum transferrin receptor and transferrin receptorferritin index identify healthy subjects with subclinical iron deficits,
Blood 92:2934-2939, 1998; reprinted with permission.)

bleeding from ulcers, gastritis due to alcohol or aspirin inges- declining body iron with increased absorption is not apparent
tion, tumors, parasitosis, diverticulitis, ulcerative colitis, or in routine laboratory test results or patient symptoms. The
hemorrhoids. In women, prolonged menorrhagia (heavy men- individual appears healthy. As the negative iron balance con-
strual bleeding) or conditions such as fibroid tumors or uterine tinues, however, a stage of iron depletion develops.
malignancies can also lead to heme iron loss. Heme iron can
also be lost excessively through the urinary tract with kidney Stage 1
stones, tumors, or chronic infections. Individuals with chronic Stage 1 of iron deficiency is characterized by a progressive loss
intravascular hemolytic processes, such as paroxysmal noctur- of storage iron. RBC development is normal, however, because
nal hemoglobinuria, can develop iron deficiency due to the the bodys reserve of iron is sufficient to maintain the transport
loss of iron in hemoglobin passed into the urine. and functional compartments through this phase. There is no
evidence of iron deficiency in the peripheral blood because
Pathogenesis RBCs survive 120 days, and the patient does not experience
Iron deficiency anemia develops slowly, progressing through symptoms of anemia. Serum ferritin levels are low, however,
stages that physiologically blend into one another but are which indicates the decline in stored iron, and this also could
useful delineations for understanding disease progression.2 As be detected in an iron stain of the bone marrow. Without
shown in Figure 19-1, iron is distributed among three compart- evidence of anemia, however, neither of these tests would be
ments: (1) the storage compartment, principally as ferritin in performed, and individuals appear healthy. The prevalence of
the bone marrow macrophages and liver cells; (2) the transport stage 1 iron deficiency in the United States has been estimated
compartment of serum transferrin; and (3) the functional at 14.4% for 1- to 2-year-olds, 3.7% for 3- to 5-year-olds, 9.3%
compartment of hemoglobin, myoglobin, and cytochromes. for 12- to 19-year-old females, and 9.2% for 20- to 49-year-old
Hemoglobin and intracellular ferritin constitute nearly 95% of females.4
the total distribution of iron.3
For a period of time as an increase in demand or increased Stage 2
loss of iron exceeds iron intake, essentially normal iron status Stage 2 of iron deficiency is defined by the exhaustion of the
continues. The body strives to maintain iron balance by accele storage pool of iron. For a time, RBC production continues as
rating absorption of iron from the intestine through a decrease normal, relying on the iron available in the transport compart-
in the production of hepcidin in the liver. This state of ment. Eventually the hemoglobin content of reticulocytes
256 PART IV Hematopathology: Erythrocyte Disorders

begins to decrease, which reflects the onset of iron deficient In this phase, the patient experiences the nonspecific symp-
erythropoiesis, but because the bulk of the circulating RBCs toms of anemia, typically fatigue and weakness, especially with
were produced during the period of adequate iron availability, exertion. Pallor is evident in light-skinned individuals but also
the overall hemoglobin measurement is still normal. Thus can be noted in the conjunctivae, mucous membranes, or
anemia is still not evident, although an individuals hemoglo- palmar creases of dark-skinned individuals.3 More severe signs
bin may begin dropping, and the RBC distribution width are not seen as often in the United States3 but include a sore
(RDW) may begin increasing as some smaller RBCs are released tongue (glossitis) due to iron deficiency in the rapidly pro
from the bone marrow. Other iron-dependent tissues, such as liferating cells of the alimentary tract and inflamed cracks at
muscles, may begin to be affected, although the symptoms may the corners of the mouth (angular cheilosis). Koilonychia
be nonspecific. The serum iron and serum ferritin levels (spooning of the fingernails) may be seen if the deficiency
decrease, whereas total iron-binding capacity (TIBC), an indi- is long-standing. Patients also may experience cravings for
rect measure of transferrin, increases (see section on iron nonfood items, called pica. The cravings may be for things such
studies). Free erythrocyte protoporphyrin (FEP), the porphyrin as dirt, clay, laundry starch, or, most commonly, ice (craving
into which iron is inserted to form heme, begins to accumulate. for the latter is called pagophagia).3
Transferrin receptors increase on the surface of iron-starved As should be evident from this discussion, numerous indi-
cells as they try to capture as much available iron as possible. viduals may be iron deficient while appearing healthy. Until
They also are shed into the plasma, so the soluble transferrin late in stage 2, they may experience no symptoms at all and are
receptor levels increase measurably. Prussian blue stain of the unlikely to come to medical attention. Even in stage 3, frankly
bone marrow in stage 2 shows essentially no stored iron, and anemic patients may not seek medical care, because the body
iron deficient erythropoiesis is evident (see subsequent descrip- is able to compensate remarkably for slowly developing anemia
tion). As in stage 1, iron deficiency in stage 2 is subclinical, and (see Chapter 18), as in the patient in the case study at the
testing is not likely to be undertaken. beginning of this chapter. Because results of routine screening
tests included in the CBC do not become abnormal until late
Stage 3 in stage 2 or early in stage 3, most patients are not diagnosed
Stage 3 of iron deficiency is frank anemia. The hemoglobin until relatively late in the progression of the iron depletion.
concentration and hematocrit are low relative to the reference
ranges. Because of the thoroughly depleted storage iron and Epidemiology
diminished transport iron, RBC precursors are unable to From the previous discussion, it is apparent that certain groups
develop normally. The number of cell divisions per precursor of individuals are more prone to develop iron deficiency
increases because hemoglobin accumulation in the developing anemia. Menstruating women are at especially high risk. Their
cells is slowed, which allows more time for divisions. The result monthly loss of blood increases their routine need for iron,
is first smaller cells with adequate hemoglobin concentration, which often is not met with the standard U.S. diet.3 For
although ultimately even these cannot be filled with hemo adolescent girls, this is compounded by increased iron needs
globin. The RBCs then become microcytic and hypochromic associated with growth. If women of childbearing age do not
(Figure 19-2). As expected, serum ferritin levels are exceedingly receive proper iron supplementation, pregnancy and nursing
low. Results of other iron studies (see later) are also abnormal, can lead to a loss of nearly 900mg of iron,5 which further
and the FEP and transferrin receptor levels continue to increase. depletes iron stores. Succeeding pregnancies can exacerbate the
problem, leading to iron deficient fetuses.5
Growing children also are at high risk.5 Growth requires
iron for the cytochromes of all new cells, myoglobin for new
muscle cells, and hemoglobin in the additional RBCs needed
to supply oxygen for a larger body. The increasing need for iron
as the child grows can be coupled with dietary inadequacies,
especially in circumstances of poverty or neglect. Cows milk
is not a good source of iron, and infants need to be placed on
iron-supplemented formula by about age 6 months, when
their fetal stores of iron become depleted.3 This assumes that
the infants were able to establish adequate iron stores by
drawing iron from their mothers in utero. Even though breast
milk is a better source of iron than cows milk,6 it is not a
consistent source.7 Therefore, iron supplementation is also
recommended for breastfed infants after 6 months of age.3
Figure 19-2 Peripheral blood film from a patient with hypochromic, micro
cytic anemia. Note the variation in red blood cell (RBC) diameters compared Iron deficiency is relatively rare in men and postmeno-
to the lymphocytes diameter. Variation in RBC diameters is termed aniso- pausal women because the body conserves iron so tenaciously,
cytosis and corresponds to an elevated RBC distribution width (RDW), a and these individuals lose only about 1mg/day. Gastrointes-
measure of variation in RBC volume. Hypochromia, microcytosis, and an tinal disease, such as ulcers, tumors, or hemorrhoids, should
elevated RDW may indicate iron deficiency anemia. be suspected in iron deficient patients in either of these groups
CHAPTER 19 Disorders of Iron and Heme Metabolism 257
if the diet is known to be adequate in iron. Regular aspirin microcytosis and hypochromia become more prominent, with
ingestion and alcohol consumption can lead to gastritis and progressively declining values for mean cell volume (MCV),
chronic bleeding. Elderly individuals, particularly those living mean cell hemoglobin (MCH), and mean cell hemoglobin
alone, may not eat a balanced diet, so pure dietary deficiency concentration (MCHC). The RBC count ultimately becomes
is seen among these individuals. In some elderly individuals, decreased, as does the hematocrit. Polychromasia may be
the loss of gastric acidity with age can impair iron absorption. apparent early, although it is not a prominent finding. A low
Iron deficiency is associated with infection by hookworms, absolute reticulocyte count confirms a diminished rate of effec-
Necator americanus and Ancylostoma duodenale. The worm tive erythropoiesis, because this is a nonregenerative anemia.11
attaches to the intestinal wall and literally sucks blood from Poikilocytosis, including occasional target cells and ellipto-
the gastric vessels. Iron deficiency is also associated with infec- cytes, may be present, although no particular shape is charac-
tion with other parasites, such as Trichuris trichiura, Schistosoma teristic or predominant. Thrombocytosis may be present,
mansoni, and Schistosoma haematobium, in which the heme iron particularly if the iron deficiency results from chronic bleeding,
is lost from the body due to intestinal or urinary bleeding. but this is not a diagnostic parameter. White blood cells
Soldiers and long-distance runners also can develop iron (WBCs) are typically normal in number and appearance. Iron
deficiency. Marching anemia develops when RBCs are hemo- deficiency should be suspected when the CBC findings show a
lyzed by foot-pounding trauma and iron is lost as hemoglobin hypochromic, microcytic anemia with an elevated RDW but no
in the urine.8 The amount lost in the urine can be so little that consistent shape changes to the RBCs.
it is not apparent on visual inspection.
Diagnosis of Iron Deficiency
Laboratory Diagnosis Iron studies remain the backbone for diagnosis of iron defi-
Iron deficiency can be readily diagnosed in later stages using ciency (see Chapter 11). They include assays of serum iron,
routine tests. Detection in the early stages requires sophisti- TIBC, transferrin saturation, and serum ferritin. Serum iron is
cated tests, but individuals are unlikely to be referred for such a measure of the amount of iron bound to transferrin (trans-
studies because there is virtually no physiologic evidence of the port protein) in the serum. TIBC is an indirect measure of
declining iron state. Nevertheless, early iron deficiency might transferrin and the available binding sites for iron in the
be suspected in an individual in a high-risk group, and appro- plasma. The percent of transferrin saturated with iron can be
priate testing can be ordered.8 The tests for iron deficiency can calculated from the total iron and the TIBC:
be grouped into three general categories: screening, diagnostic,
and specialized. serum iron (g dL ) 100
Transferrin saturation (% sat ) =
TIBC (g dL )
Screening for Iron Deficiency Anemia
When iron deficient erythropoiesis is under way, the CBC Ferritin is not truly an extracellular protein, because it pro-
results begin to show evidence of anisocytosis, microcytosis, vides an intracellular storage repository for metabolically active
and hypochromia (see Figure 19-2). The classic picture of iron iron. However, ferritin is present in serum, and serum levels
deficiency anemia in stage 3 includes a decreased hemoglobin reflect the levels of iron stored within cells. Serum ferritin is an
level. An RDW greater than 15% is expected and may precede easily accessible surrogate for stainable bone marrow iron. The
the decrease in hemoglobin.9 For patients in high-risk groups, iron studies are used collectively to assess the iron status of an
the elevated RDW can be an early and sensitive indicator of individual. Table 19-1 shows that, as expected, serum ferritin
iron deficiency.10 As the hemoglobin level continues to fall, and serum iron values are decreased in iron deficiency anemia.

TABLE 19-1 Results of Iron Studies in Microcytic, Hypochromic Anemias


Anemia of Chronic Sideroblastic
Iron Deficiency Thalassemia Minor Inflammation Anemia Lead Poisoning
Serum ferritin /N /N N
Serum iron /N /N Variable
TIBC N /N N
Transferrin saturation /N /N
FEP/ZPP N (marked)
BM iron (Prussian No stainable iron /N /N N
blue reaction)
Sideroblasts in BM None N None/very few (ringed) N (ringed)
Other special tests Hb A2 (-thalassemia Specific tests for inflammatory ALA in urine
minor) disorders or cancer Whole-blood lead levels

, Increased; , decreased; ALA, aminolevulinic acid; BM, bone marrow; FEP/ZPP, free erythrocyte protoporphyrin/zinc protoporphyrin; Hb, hemoglobin; N, normal.
258 PART IV Hematopathology: Erythrocyte Disorders

Transferrin levels increase when the hepatocytes detect low


iron levels, and research shows that this is a transcriptional and
posttranslational response to low iron levels.12 The result is a
decline in the iron saturation of transferrin that is more dra-
matic than might be expected simply from the decrease in
serum iron level.
It is important that specimens for iron studies are drawn
while fasting and early in the morning when levels are highest.
Iron levels show a diurnal variation, with levels dropping
throughout the day.13 Iron absorbed from a meal can falsely
elevate levels.14
The amount of hemoglobin in reticulocytes can be assessed
on some automated hematology analyzers.15 The reticulocyte
hemoglobin value is analogous to the MCH, but for reticulo-
cytes. The MCH is the average weight of hemoglobin per cell Figure 19-3 Bone marrow smear for a patient with iron deficiency
across the entire RBC population. Some of the RBCs are nearly anemia. The late-nucleated red blood cells show the characteristic shaggy
120 days old, whereas others are just 1 to 2 days old. If iron blue cytoplasm due to asynchrony in maturation (1000). (Courtesy Ann Bell,
deficiency is developing, the MCH does not change until a University of Tennessee, Memphis.)
substantial proportion of the cells are iron deficient, and the
diagnosis is effectively delayed for weeks or months after iron Treatment and Its Effects
deficient erythropoiesis begins. The reticulocyte hemoglobin Treatment
value is able to assess iron deficient erythropoiesis within days The first therapy for iron deficiency is to treat any underlying
as the first iron deficient cells leave the bone marrow. It is a contributing cause, such as hookworms, tumors, or ulcers. As
sensitive indicator of iron deficiency. Even in stage 2 of iron in the treatment of simple nutritional deficiencies or increased
deficiency, before anemia is apparent, the reticulocyte hemo- need, dietary supplementation is necessary to replenish the
globin will be low.15 bodys iron stores. Oral supplements of ferrous sulfate (3
tablets/day containing 60mg of elemental iron) are the stan-
Specialized Tests dard prescription.3 The supplements should be taken on an
Other tests, although not commonly used for the diagnosis of empty stomach to maximize absorption. Many patients experi-
iron deficiency, show abnormalities that become important in ence side effects such as nausea and constipation, however,
the differential diagnosis of similar conditions. Test results for which leads to poor patient adherence. Vigilance on the part
the accumulated porphyrin precursors to heme are elevated of the health care providers is important to ensure that patients
(see Table 19-1). FEP accumulates when iron is unavailable. In complete the course of iron replacement, which usually lasts 6
the absence of iron, FEP may be preferentially chelated with months or longer.18 In rare cases in which intestinal absorption
zinc to form zinc protoporphyrin (ZPP).16 The FEP and zinc of iron is impaired, as with gastric achlorhydria, parenteral
chelate can be assayed fluorometrically. Soluble transferrin administration of iron dextrans can be used, although the side
receptors (sTfR) also can be assayed using immunoassay. Levels effects of this therapy are notable.3 Because of the risks of blood
increase as the disease progresses and individual cells seek to transfusions, they are rarely warranted for the correction of
take in as much iron as possible.17 uncomplicated iron deficiency unless the patients hemoglobin
A bone marrow assessment is not indicated for suspected level has become dangerously low.
uncomplicated iron deficiency. A therapeutic trial of iron (see
Response to Treatment) provides a less invasive and less expen- Response to Treatment
sive diagnostic assessment. Marrow examination may be per- When optimal treatment with iron is initiated, the effects are
formed, however, if the diagnosis is complicated or if other test quickly evident. Reticulocyte hemoglobin will correct within 2
results are equivocal. With routine stains, the iron deficient days.15 Reticulocyte counts (relative and absolute) begin to
bone marrow appears hyperplastic early in the progression of increase within 5 to 10 days.19 The anticipated rise in hemo-
the disease, with a decreased myeloid-to-erythroid ratio as a globin appears in 2 to 3 weeks, and levels should return to
result of increased erythropoiesis.3 As the disease progresses, normal for the individual by about 2 months after the initia-
hyperplasia subsides, and the profound deficiency of iron leads tion of adequate treatment.19 The peripheral blood film and
to slowed RBC production. Polychromatic normoblasts (i.e., indices still reflect the microcytic RBC population for several
rubricytes) show the most dramatic morphologic changes months, but the normocytic population eventually predomi-
(Figure 19-3). Nuclear-cytoplasmic asynchrony is evident, with nates. Iron therapy must continue for another 3 to 4 months
cytoplasmic maturation lagging behind nuclear maturation. to replenish the storage pool and prevent a relapse.
Without the pink provided by hemoglobin, the cytoplasm If the patient has been adherent to the therapy regimen, the
remains bluish after the nucleus has begun to condense. The failure to respond to iron treatment points to the need for
cell membranes appear irregular and are usually described as further investigation. The patient may be iron deficient but
shaggy. experiencing continued occult loss of blood or inadequate
CHAPTER 19 Disorders of Iron and Heme Metabolism 259

absorption. Alternatively, causes of hypochromic, microcytic A second acute phase reactant seems to contribute to anemia
anemia unrelated to iron deficiency, such as thalassemia, of chronic inflammation, although probably to a much smaller
should be investigated. extent than hepcidin. Lactoferrin is an iron-binding protein in
the granules of neutrophils. Its avidity for iron is greater than
that of transferrin. Lactoferrin is important intracellularly for
ANEMIA OF CHRONIC INFLAMMATION
phagocytes to prevent phagocytized bacteria from using intra-
Anemia is commonly associated with systemic diseases, includ- cellular iron for their metabolic processes.26 During infection
ing chronic inflammatory conditions such as rheumatoid and inflammation, however, neutrophil lactoferrin also is
arthritis, chronic infections such as tuberculosis or human released into the plasma. There it scavenges available iron, at
immunodeficiency virus infection, and malignancies. Cart- the expense of transferrin. When it is carrying iron, the lactofer-
wright20 was the first to suggest that although the underlying rin is bound to macrophages and liver cells that salvage the
diseases seem quite disparate, the associated anemia may be iron. RBCs are deprived of this iron, however, because they do
from a single cause, proposing the concept of anemia of not have lactoferrin receptors.
chronic disease. This anemia represents the most common Finally, a third acute phase reactant, ferritin, contributes to
anemia among hospitalized patients.21 the anemia of chronic inflammation. Increased levels of ferritin
in the plasma also bind some iron. Because developing RBCs
Etiology do not have a ferritin receptor, this iron is unavailable for
Although the anemia associated with chronic systemic dis incorporation into hemoglobin.
orders was originally called anemia of chronic disease, chronic The result of these effects is that, although iron is present
blood loss is not among the conditions leading to the anemia in abundance in bone marrow macrophages, its release to
of chronic disease. Chronic blood loss results in clear-cut iron developing erythrocytes is slowed. This can be seen histologi-
deficiency. Anemia of chronic disease is more correctly termed cally with iron stains that show iron in macrophages but not
anemia of chronic inflammation, because inflammation is the in erythroblasts.27 The effect on the developing RBCs is essen-
unifying factor among the three aforementioned general types tially no different from that in mild iron deficiency, because
of conditions in which this anemia is seen. The central feature they are effectively deprived of the iron.
of anemia of chronic inflammation is sideropenia in the face Diminished erythropoiesis and a blunted response to eryth-
of abundant iron stores. The cause is now understood to be ropoietin also may contribute to the anemia of chronic inflam-
largely impaired ferrokinetics. mation.28 The impaired ferrokinetics, however, are likely the
The apparent inconsistency of decreased serum iron but more significant cause of the anemia.
abundant iron stores is explained by the role of hepcidin in
regulation of body iron. Hepcidin is a hormone produced by Laboratory Diagnosis
hepatocytes to regulate body iron levels, particularly absorption The peripheral blood picture in anemia of chronic inflamma-
of iron in the intestine and release of iron from macrophages. tion is that of a mild anemia with hemoglobin concentration
Hepcidin interacts with the transmembrane protein ferroportin, usually 9 to 11g/dL and without reticulocytosis. The cells are
which exports iron from enterocytes into the plasma, reducing usually normocytic and normochromic, although microcytosis
the amount of iron absorbed into the blood from the intestine.22 and hypochromia develop in about one third of patients and
Macrophages and hepatocytes also use ferroportin to export iron may represent coexistent iron deficiency.3 The inflammatory
into plasma and are affected by hepcidin.22,23 When body iron condition leading to the anemia also may cause leukocytosis,
levels decrease, hepcidin production by hepatocytes decreases,24 thrombocytosis, or both. Iron studies (see Table 19-1) show
and enterocytes export more iron into the plasma. Macrophage low serum iron and TIBC values. Because hepatocyte produc-
and hepatocyte release of iron also increases. When iron levels tion of transferrin is regulated by iron levels,12 the low TIBC
are high, hepcidin increases, enterocytes export less iron into (an indirect measure of transferrin) reflects the abundant iron
the plasma, and macrophages and hepatocytes retain iron. stores in the body. The transferrin saturation may be normal
Hepcidin is an acute phase reactant,25 so levels increase or low. Serum ferritin level is usually increased beyond the
during inflammation, regardless of iron levels in the body. As value that would be expected for the same patient in the
a result, during inflammation, there is a decrease in iron absence of the inflammatory condition. It may not be outside
absorption from the intestine and iron release from macro- the reference range, but it is nevertheless increased. The failure
phages and hepatocytes. Although there is plenty of iron in the to incorporate iron into heme results in elevation of FEP,
body, it is unavailable to developing RBCs because it is seques- although this test typically is not used diagnostically. The bone
tered in the macrophages and hepatocytes. marrow shows hypoproliferation of the RBCs, consistent with
This response of hepcidin during inflammation is likely a the lack of reticulocytes in the peripheral blood. Prussian blue
nonspecific defense against invading bacteria. If the body can stain of the bone marrow confirms abundant stores of iron in
sequester iron, it reduces the amount of iron available to bac- macrophages, although not in RBC precursors, but bone
teria and contributes to their demise. Although this response marrow examination is not usually required in the diagnostic
of hepcidin is not harmful during disorders of short duration, evaluation.
chronically high levels of hepcidin embargo iron for long Patients with iron deficiency anemia who have an inflam-
periods, which leads to diminished production of RBCs. matory condition present a special diagnostic dilemma. The
260 PART IV Hematopathology: Erythrocyte Disorders

iron deficiency may be missed because of the increase in serum which glycine is condensed with succinyl coenzyme A to form
ferritin levels associated with the inflammation. Iron deficiency aminolevulinic acid.
anemia and anemia of chronic inflammation can be distin-
guished in such situations by measuring serum (soluble) trans- Lead Poisoning
ferrin receptors.29 These receptors are sloughed from cells into The acquired conditions leading to sideroblastic anemia
the plasma. As noted earlier, levels increase during iron defi- constitute a diverse group in themselves. Certain drugs, such
ciency anemia but remain essentially normal during anemia of as chloramphenicol or isoniazid, can induce sideroblastic
chronic inflammation. anemia.33 Other toxins, including heavy metals, also have been
implicated. Among these, lead is a significant public health
Treatment concern. Adults may be exposed at work to leaded compounds.
Therapeutic administration of erythropoietin can correct Adults and children living in older homes can be exposed to
anemia of chronic inflammation,30 but iron must be adminis- lead from paints produced before the 1970s. They are at risk if
tered concurrently, because stored body iron remains seques- dust is created during renovations. Toddlers and crawling
tered and unavailable.31 The anemia is typically not severe, infants are at special risk from getting dust on their hands and
however, and this costly treatment is warranted only in select placing them in their mouths. Although anyone can experience
patients. The best course of treatment is effective control or lead poisoning, it is of special concern in children, because
alleviation of the underlying condition. the metal affects the central nervous system and the hemato-
logic system, leading to impaired mental development.34 In
children and adults with lead poisoning, a peripheral neuropa-
SIDEROBLASTIC ANEMIAS
thy35 can be seen with abdominal cramping and vomiting or
Just as anemia can result from inadequate supplies of iron for seizures.
production of hemoglobin, diseases that interfere with the Lead interferes with porphyrin synthesis at several steps. The
production of adequate amounts of heme also can produce most critical are as follows (see Figure 10-5):
anemia. (See Chapter 10 for a discussion of heme synthesis.) 1. The conversion of aminolevulinic acid to porphobilinogen;
As in iron deficiency, the anemia may be microcytic and hypo- the result is the accumulation of aminolevulinic acid.
chromic. In contrast to iron deficiency, however, iron is abun- 2. The incorporation of iron into protoporphyrin IX by fer-
dant in the bone marrow. A Prussian blue stain of the bone rochelatase (also called heme synthetase); the result is accu-
marrow shows normoblasts with iron deposits in the mito- mulation of iron and protoporphyrin.36
chondria surrounding the nucleus. Its presence in the mito- Of these, the impairment of ferrochelatase is probably the
chondria shows that the iron is awaiting incorporation into more significant. Accumulated aminolevulinic acid is measur-
heme. These ringed sideroblasts are the hallmark of the sidero- able in the urine, and protoporphyrin is measurable in an
blastic anemias (Figure 19-4). extract of RBCs as FEP or ZPP.
The sideroblastic anemias are a diverse group of diseases Anemia, when present in lead poisoning, is most often
including hereditary and acquired conditions (Box 19-1). normocytic and normochromic; however, with chronic expo-
Among the hereditary forms, X-linked and autosomal varieties sure to lead, a microcytic, hypochromic clinical picture may be
of this condition are known. Some patients experience at least seen. The degree of anemia in adults may not be dramatic, but
modest improvement of anemia with pharmacologic doses of in children it may be more profound. The reticulocyte count
pyridoxine to stimulate heme synthesis.32 Pyridoxine is a cofac- in acute poisoning may be quite elevated, which suggests that
tor in the first step of porphyrin synthesis (see Figure 10-5) in

BOX 19-1 Disorders Included in Sideroblastic


Anemias

Hereditary
X-linked
Autosomal

Acquired
Primary sideroblastic anemia (refractory)
Secondary sideroblastic anemias caused by drugs and bone marrow
toxins
Antitubercular drugs
Chloramphenicol
Alcohol
Lead
Figure 19-4 Ringed sideroblasts (arrows) in bone marrow shown with Chemotherapeutic agents
Prussian blue stain (1000).
CHAPTER 19 Disorders of Iron and Heme Metabolism 261

TABLE 19-2 Erythropoietic Porphyrias


Congenital Erythropoietic Porphyria (CEP) Erythropoietic Protoporphyria (EPP)
Enzyme deficiency Uroporphyrinogen III synthase Ferrochelatase
Affected gene UROS FECH
Inheritance Autosomal recessive Autosomal dominant
Clinical features Photosensitivity, hemolytic anemia Photosensitivity; anemia is mild if present
Laboratory features
Red blood cells Protoporphyrin N
Uroporphyrin N
Coproporphyrin N
Urine Porphobilinogen N N
Uroporphyrin N
Coproporphyrin N
Feces Protoporphyrin N
Coproporphyrin N
Confirmatory tests Uroporphyrinogen III synthase activity Ferrochelatase activity
Genetic testing Genetic testing

Adapted from Desnick RJ, Astrin KH: The porphyrias. In Fauci AS, Braunwald E, Kasper DL, etal: Harrisons principles of internal medicine, ed 17, New York, 2008,
McGraw Hill, chap 352. Available at: http://www.accessmedicine.com/content.aspx?aID=2883658. Accessed June 19, 2010.
, Minimally increased levels; , moderately increased levels; , markedly increased levels; moderately decreased; markedly decreased.

the anemia has a hemolytic component. The presence of a When an enzyme in heme synthesis is missing, the products
hemolytic component is supported by studies showing impair- from earlier stages in the pathway accumulate in cells that
ment of the pentose-phosphate shunt by lead,37 which makes actively produce heme proteins, such as erythrocytes and hepa-
the cells sensitive to oxidant stress as in glucose-6-phosphate tocytes. The excess leaks from the cells as they age or die, and
dehydrogenase deficiency (see Chapter 23). In contrast, chronic may be excreted in urine or feces, which allows diagnosis. The
poisoning results in hypoplasia of the marrow.38 Basophilic accumulated products also deposit in body tissues. Some of
stippling is a classic finding associated with lead toxicity. Lead the accumulated products are fluorescent. Their deposition in
inhibits pyrimidine 5-nucleotidase, an enzyme involved in the skin can lead to photosensitivity with severe burns upon expo-
breakdown of ribosomal ribonucleic acid (RNA) in reticulo- sure to sunlight. Accumulation during childhood leads to fluo-
cytes.39 This causes undegraded ribosomes to aggregate, forming rescence of developing teeth and bones. Only two of the
basophilic stippling. Because basophilic stippling is also seen porphyrias have hematologic manifestations; the others have
in other anemias, this is not a pathognomonic finding. The size a greater effect on liver cells. Even in those with hematologic
of the aggregates in lead poisoning is typically large, so that the effects, the hematologic impact is relatively minimal, and pho-
stippling is heavier than that seen in many anemias and thus tosensitivity is a greater clinical problem. The fluorescence of
represents truly punctate basophilia. the accumulated compounds can be used diagnostically as
Removal of the drug or toxin is usually successful for the described earlier for FEP. In a sample of bone marrow, the
treatment of acquired sideroblastic anemias. In the case of lead, erythroblasts will be bright red under a fluorescent microscope.
salts of ethylenediaminetetraacetic acid are often used to Table 19-2 summarizes the deficient enzymes, affected genes,
chelate the lead present in the body so that it can be excreted inheritance, and clinical and laboratory features that can be
in the urine.38 used in the laboratory diagnosis of the hematologically signifi-
cant porphyrias.
Porphyrias
Lead poisoning is an example not only of an acquired sidero-
IRON OVERLOAD
blastic anemia, but also of an acquired porphyria. The porphyr-
ias are diseases characterized by impaired production of heme. Chapter 11 describes the bodys tenacity in conserving iron.
The impairments to heme synthesis may be acquired, as with For some individuals, this tenacity becomes the basis for
lead poisoning, or hereditary. The term porphyria is most often disease related to excess iron accumulations in nearly all cells.
used to refer to the hereditary conditions. Among the inherited Iron overload may be primary as in hereditary hemochroma-
disorders, single deficiencies of most enzymes in the synthetic tosis or secondary to chronic anemias and their treatments. In
pathway for heme have been identified (see Figure 10-5). both cases, the toxic effects of excess iron lead to serious health
Although even the autosomal dominant conditions are rela- problems.
tively rare, the disease has been influential historically. The
intermarrying European monarchies of past centuries were Etiology
plagued with some variants of the porphyrias in which psycho- Excess accumulation of iron results from acquired or hereditary
sis is a prominent clinical feature. conditions in which the bodys rate of iron acquisition exceeds
262 PART IV Hematopathology: Erythrocyte Disorders

the rate of loss, which is usually about 1mg/day. Regardless hereditary hemochromatosis, which is the excess absorption of
of the source of the iron, the bodys first reaction is to store dietary iron. Enterocytes do not absorb iron from the diet
excess iron in the form of ferritin and, ultimately, hemosiderin through a transferrin-dependent mechanism45; rather, it is
within cells. Eventually the storage system is overwhelmed, absorbed by the divalent metal transporter 1 on the luminal
and, as described later, parenchymal cells are damaged in side of enterocytes.46 The active transport of iron into the
organs including the liver, heart, and pancreas. plasma seems to be crucial in the pathophysiology of heredi-
Accumulation of excess iron may be an acquired condition. tary hemochromatosis. This is accomplished by ferroportin on
It occurs when there is a need for repeated transfusions, as in the basolaminal side of the enterocyte. In hereditary hemo-
the treatment of anemias such as thalassemia. The iron present chromatosis, the enterocytes may absorb no more iron than
in the transfused RBCs exceeds the usual 1mg/day of iron normal from the diet, but they transport more of it into the
typically added to the bodys stores by a healthy diet. This is plasma. Because hepcidin regulates this process, it plays a
called transfusion-related hemosiderosis. central role in the pathogenesis of hereditary hemochromato-
Hemochromatosis may develop as a result of mutations sis. In hepatocytes, iron-bound transferrin, HFE, transferrin
affecting the proteins of iron metabolism. An autosomal reces- receptor 2, and hemojuvelin regulate hepcidin expression.46
sive disease has been recognized for many years; however, only How these interactions affect hepcidin transcription and
more recently have advances in molecular biology allowed produce the hereditary hemochromatosis phenotype has yet to
identification of the mutant genes that contribute to the phe- be fully determined. Still, a central role for hepcidin is sup-
notype. Homozygous hereditary hemochromatosis involving ported by the discovery that mutations in the hepcidin gene
certain genes occurs in approximately 5 of 1000 northern itself, HAMP, or mutations in HJV (HFE2), the gene that codes
Europeans.40 Heterozygosity approaches 13%.40 The first two for hemojuvelin, can produce a disease complex affecting chil-
mutations known to produce the hereditary hemochromatosis dren that is identical to hereditary hemochromatosis.46 Muta-
phenotype involve HFE, a gene on the short arm of chromo- tions of genes coding for ferroportin and transferrin receptor
some 6 that encodes an HLA class Ilike molecule that is can also produce a phenotype similar to that of hereditary
closely linked to HLA-A.41 The most common of the two hemochromatosis.46 What has emerged is a picture of heredi-
mutations substitutes tyrosine for cysteine at position 282 tary hemochromatosis as a general phenotype that can be pro-
(Cys282Tyr), and the other substitutes aspartate for histidine duced by various genotypes when the gene for any of the iron
at position 63 (His63Asp).42 The normal HFE protein binds regulatory proteins is mutated (Table 19-3). As noted earlier,
2-microglobulin intracellularly.42 This binding is necessary for the roles and interactions of all these proteins in normal iron
the HFE to appear on the cell surface, where it interacts with regulation have not been fully elucidated, and a completely
transferrin receptor 1. Interaction of HFE with transferrin satisfactory explanation of the pathogenesis of hereditary
receptor 1 reduces transferrin binding to the receptor, inhibit- hemochromatosis has yet to be established. Suffice it to say
ing cellular iron absorption.43 The mutated HFE molecule that because the biologic default is to absorb and store iron,
either does not bind 2-microglobulin and thus does not reach and the regulatory mechanisms typically dampen that process,
the cell surface, or does not bind the transferrin receptor 1 if failure of normal regulation due to mutations leads to excessive
it does reach the cell surface.44 In either case, the result is that, absorption and storage.
when HFE is mutated, the transferrin receptor 1 is inappropri-
ately available for transferrin binding, even when the cell is Pathogenesis
replete with iron. The processes described previously lead to increased amounts
The previously described role of HFE in cellular iron uptake of iron in parenchymal cells throughout the body. The first
does not address a significant feature in the pathogenesis of cellular reaction to excess iron is to form hemosiderin,

TABLE 19-3 Known Mutations Producing Phenotypes Similar to Hereditary Hemochromatosis Phenotypes
TfR2-Related
HFE-Related Hereditary Ferroportin-Related
Feature Hemochromatosis Juvenile Hereditary Hemochromatosis Hemochromatosis Iron Overload
Affected gene HFE HAMP HJV (HFE2) TfR2 SLC40A1
Mutated protein HFE Hepcidin Hemojuvelin Transferrin receptor 2 Ferroportin
Normal function of Inhibits TfR1-mediated Downregulates Regulates hepcidin Provides hepatocyte iron Transports iron out of
affected protein iron uptake; regulates ferroportin-mediated iron expression uptake; regulates enterocytes and
hepcidin expression transport in macrophages hepcidin expression macrophages
and enterocytes
Age of onset of 30-40 Teens-20 Teens-20 20-40, mild 30-40
symptoms (yr)

TfR1, Transferrin receptor 1; TfR2, transferrin receptor 2.


CHAPTER 19 Disorders of Iron and Heme Metabolism 263

essentially a degenerate and nonmetabolically active form of Laboratory Diagnosis


ferritin. When cells exhaust the capacity to store iron as hemo- Laboratory testing in hemochromatosis serves three purposes.
siderin, free iron (ferrous) accumulates intracellularly. In the It can be used to screen for the condition, diagnose the cause
presence of oxygen, ferrous iron initiates the generation of of organ damage, and monitor treatment. Transferrin satura-
superoxide and other free radicals, which results in the peroxi- tion (which is increased in the disease) is the common screen-
dation of membrane lipids.3,47 The membranes affected include ing test for hereditary hemochromatosis51 and can be performed
not only the cell membranes, but also mitochondrial, nuclear, cost-effectively in populations with a disease prevalence of at
and lysosomal membranes. Cell respiration is compromised, least 3 per 1000.52 In a screening situation, transferrin satura-
and lysosomal enzymes are released intracellularly. Vitamins E tion of 60% or greater on repeated tests warrants further inves-
and C can act to moderate the effects and interrupt the chain tigation.51 Transferrin saturation also can be used to follow
reaction, but in iron overload, even these protective mecha- patients with transfusion-dependent anemia to detect the
nisms are overwhelmed. The ultimate result is cell death due development of hemosiderosis.
to irreversible membrane damage. Individuals with undiagnosed hereditary hemochromatosis
Because all cells except mature RBCs require iron and have may come to medical attention because of organ function
the cellular machinery for iron acquisition, most cells have the problems, or the disease may be discovered incidentally with
potential for iron damage. The tissues most obviously affected routine laboratory testing. Abnormal results on common tests
are the skin, where deposition of hemosiderin gives the skin a of liver function (e.g., elevated alanine transaminase levels)
golden color; the liver, where cirrhosis-induced jaundice and may be among the first laboratory findings that lead a physi-
subsequent cancer develop; and the pancreas, where damage cian to order further testing to identify the cause. Because
results in diabetes mellitus. Hence the traditional characteriza- inflammation is minimal, however, a finding of diminished
tion of hemochromatosis is bronzed diabetes. The heart levels of the livers synthetic products, such as albumin, may
muscle also is especially vulnerable to excessive iron deposi- be more helpful. If hereditary hemochromatosis is among the
tion, which leads to congestive heart failure. Early diagnosis disorders being considered to explain organ dysfunction,
and treatment (see later) can prevent the development of these serum iron, transferrin saturation, and serum ferritin testing is
secondary effects of iron overload. Hepatocellular carcinoma warranted. Elevations in these values are among the earliest
occurs more frequently in patients with hemochromatosis. findings in most forms of hemochromatosis. Genetic testing
Mutations of the p53 tumor suppressor gene seem to contribute for known mutations provides confirmation of the diagnosis
to the pathogenesis of the carcinoma,48 and there is some evi- for most patients with hereditary hemochromatosis.
dence that the free radicals produced by the iron cause the Whether hemochromatosis is acquired or hereditary, serum
mutations in the p53 gene.49 ferritin level provides an assessment of the degree of iron over-
The development of clinical disease associated with excess load and can be monitored after treatment is initiated to reduce
iron is heavily influenced by other physiologic conditions and iron stores. Hemoglobin concentration and hematocrit are
environment. The phenotypic expression of the tissue damage inexpensive tests that can also be used to monitor treatment as
described earlier in hereditary hemochromatosis is more described later.
common in men, although the gene frequency is not higher in Determination of the actual extent of tissue damage is
men. This is because the blood loss associated with menstrua- beyond the scope of the clinical laboratory. Liver biopsy with
tion and childbirth forestalls the effects of excess iron in assessment of iron staining and degree of scarring in liver speci-
affected women, and they usually develop clinical symptoms mens is essential to determining the degree of organ damage.
later in life than affected men. In each sex, homozygous indi-
viduals develop disease faster than heterozygotes. The amount Treatment
of iron available in the diet for absorption affects the rate at The treatment of secondary tissue damage, such as liver cirrho-
which disease can develop. Substances that can promote iron sis and heart failure, follows standard protocols. Treatment of
absorption even in normal individuals, such as ascorbic acid the underlying condition leading to excess iron accumulation
and alcohol, also affect absorption in individuals with hemo- is also needed. Hereditary hemochromatosis and transfusion-
chromatosis. Finally, time is an important factor in the devel- related hemosiderosis require different treatment approaches.
opment of disease. In classic hereditary hemochromatosis, In forms of hereditary hemochromatosis, withdrawal of blood
individuals usually harbor 20 to 30g of iron by the time their by phlebotomy provides a simple, inexpensive, and effective
disease becomes clinically evident at the age of 40 to 60 years.50 means of removing iron from the body. The regimen calls
This is more than 10 times the amount of stored iron in normal for weekly phlebotomy early in treatment to remove about
individuals and represents just 1 to 2mg/day of excess iron 500mL of blood per treatment. Maintenance phlebotomies
absorbed over many years.51 In the juvenile form of the disease are required about every 3 months for life.3 Hemoglobin levels
associated with mutations to the hepcidin or hemojuvelin are monitored, and a mild anemia is sought and maintained.
genes, the process of iron accumulation is accelerated, so that Such monitoring is an easy and inexpensive substitute for iron
these effects may appear as early as the teenage years.46 In studies because, as explained in the discussion of iron defi-
transfusion-related hemosiderosis, the frequency of transfu- ciency, iron stores must be exhausted before anemia develops.
sions over time affects the rate of development of clinical Individuals who rely on transfusions to maintain hemo
disease. globin levels and prevent anemia cannot be treated with
264 PART IV Hematopathology: Erythrocyte Disorders

phlebotomy. Instead, iron-chelating drugs are used to bind with its bound iron, it is readily excreted in the urine. Recently,
excess iron in the body for excretion. Desferrioxamine is the oral iron chelators have been developed.53,54 Although they also
classic treatment, although it is not without side effects. The have side effects, the convenience of oral administration with
drug typically is injected subcutaneously to maximize exposure potential for improved patient outcomes may lead to a greater
time for iron binding.3 When absorbed into the bloodstream reliance on this form of treatment.

SUMMARY
Impaired iron or heme metabolism can result in microcytic, hypo pathway are deficient or impaired. Deficiencies of these enzymes
chromic anemias. may be hereditary, as in the porphyrias, or acquired, as in heavy
Three conditions affecting iron metabolism can result in micro metal poisoning. The most common of the latter conditions is lead
cytic, hypochromic anemias: iron deficiency, anemia of chronic poisoning.
inflammation, and sideroblastic anemias, especially lead poison Iron studies in sideroblastic anemias show elevated levels of
ing. The RBCs in thalassemias also may be microcytic and hypo serum iron and serum ferritin, variable TIBC, and increased trans
chromic, and this condition must be differentiated from the ferrin saturation. Test values for the accumulating products of the
anemias of disordered iron metabolism. heme synthetic pathway, such as ZPP or FEP, are also elevated.
Iron deficiency results from inadequate iron intake, increased Lead interferes with several steps in heme synthesis, preventing
need, decreased absorption, or excessive loss. All four of these iron incorporation into heme and resulting in a normocytic, normo
situations create a relative deficit of body iron that over time chromic anemia, although with long-term exposure it can be micro
results in a microcytic, hypochromic anemia. cytic and hypochromic. Lead also impairs glucose-6-phosphate
Infants, children, and women of childbearing age are at greatest dehydrogenase, which adds a hemolytic component to the anemia.
risk for iron deficiency anemia. If iron deficiency anemia is present Children are especially vulnerable to the effects of lead on the
in men and postmenopausal women, the possibility of gastro central nervous system, which may result in irreversible brain
intestinal bleeding should be investigated, because it is the damage. Treatment consists of removing the source of lead from
primary, although not the only, cause of iron loss. the patients environment and, if necessary, chelating drug therapy
Iron deficiency is suspected when the CBC shows microcytic, to facilitate excretion of lead in the urine.
hypochromic RBCs and elevated RDW, but no consistent morpho Because the body has no mechanism for iron excretion, iron
logic abnormality. The diagnosis is confirmed with iron studies overload can occur when transfusions are used to sustain patients
showing low levels of serum iron and serum ferritin, elevated TIBC, with chronic anemias such as thalassemia (called transfusion-
and decreased transferrin saturation. related hemosiderosis).
Iron deficiency is treated by oral supplementation, and with good A defective HFE gene can lead to hereditary hemochromatosis by
patient adherence, the anemia should be corrected within 3 causing all body cells to bind transferrin inappropriately. Affected
months. Gastrointestinal distress resulting from iron supplements men develop symptoms earlier in life than women; homozygotes
can make patient adherence a significant concern. develop more severe disease than heterozygotes.
The anemia of chronic inflammation is associated with chronic Mutations of other genes affecting iron regulation can produce a
infections such as tuberculosis, chronic inflammatory conditions phenotype similar to that of hereditary hemochromatosis. When
such as rheumatoid arthritis, and tumors. It may be a micro the hepcidin or hemojuvelin gene is mutated, the disease develops
cytic, hypochromic anemia, but most often is normocytic, early in life, affecting even teenagers.
normochromic. Free iron becomes available in cells when ferritin and hemosiderin
Increased levels of hepcidin, an acute phase reactant, decrease become saturated. Free iron causes tissue damage by creating
iron absorption in the intestines and sequester iron in macrophages free radicals that lead to cell membrane damage and perhaps
and hepatocytes in the anemia of chronic inflammation. Bone mutations. The liver, pancreas, skin, and heart muscle are espe
marrow macrophages show abundant stainable iron, whereas cially damaged by excess iron deposition.
developing RBCs show inadequate iron. Inflammatory cellular Transferrin saturation is a good screening test for hemochroma
products also impair the production and action of erythropoietin. tosis and is elevated in those with the condition. Hereditary hemo
Iron studies in the anemia of chronic inflammation show decreased chromatosis and the other known forms of the disease can be
serum iron level, decreased TIBC, decreased transferrin satura diagnosed using genetic testing to identify mutated genes.
tion, and normal or increased ferritin level. Hereditary hemochromatosis and similar diseases are treated by
Sideroblastic anemias develop when the synthesis of protoporphy lifelong periodic phlebotomy to induce a mild iron deficiency
rin or the incorporation of iron into protoporphyrin is blocked. The anemia and keep body iron levels low. Transfusion-related hemo
result is accumulation of iron in the mitochondria of developing siderosis must be treated with iron-chelating drugs, such as
RBCs. When stained using Prussian blue, the iron appears in desferrioxamine.
deposits around the nucleus of the developing cells. These cells
are called ringed sideroblasts. Now that you have completed this chapter, go back and
Protoporphyrin synthesis and iron incorporation into protoporphy read again the case study at the beginning and respond
rin can be blocked when any of the enzymes of the heme synthetic to the questions presented.
CHAPTER 19 Disorders of Iron and Heme Metabolism 265

R E V I E W Q UESTIONS
1. The mother of a 4-month-old infant who is being breast- 4. A 35-year-old white woman went to her physician
fed sees her physician for a routine postpartum visit. She complaining of headaches, dizziness, and nausea. The
expresses concern that she may be experiencing postpar- headaches had been increasing in severity over the last 6
tum depression because she does not seem to have any months. This was coincident with her move into an older
energy. Although the physician is sympathetic to the house built about 1900. She had been renovating the
patients concern, she orders a CBC and iron studies house, including stripping paint from the woodwork. Her
seeking an organic explanation for the patients symptoms. CBC results showed a mild hypochromic, microcytic
The results are as follows: anemia with polychromasia and basophilic stippling
CBC: all results within reference range except noted. Which of the following tests would be most useful
RDW = 15% in confirming the cause of her anemia?
Serum iron: decreased a. Serum lead level
TIBC: increased b. Serum iron level and TIBC
% transferrin saturation: decreased c. Absolute reticulocyte count
Serum ferritin: decreased d. Prussian blue staining of the bone marrow to detect
Correlate the patients laboratory and clinical findings. iron stores in macrophages
What can you conclude?
a. The results of the iron studies reveal findings 5. In men and postmenopausal women whose diets are
consistent with a thalassemia that was apparently adequate, iron deficiency anemia most often results
previously undiagnosed. from:
b. The patient is in stage 2 of iron deficiency, before a. Increased need associated with aging
frank anemia develops. b. Impaired absorption in the gastric mucosa
c. The results of the iron studies are inconsistent with c. Chronic gastrointestinal bleeding
the CBC results, and a laboratory error should be d. Diminished resistance to hookworm infections
suspected.
d. There is no evidence of a hematologic explanation for 6. Which of the following individuals is at greatest risk for
the patients symptoms. development of iron deficiency anemia?
a. A 15-year-old boy who eats mainly fast food and junk
2. A bone marrow biopsy was performed as part of the food
cancer staging protocol for a patient with Hodgkin b. A 37-year-old woman who has never been pregnant
lymphoma. Although no evidence of spread of the tumor and has amenorrhea
was apparent in the marrow, other abnormal findings c. A 63-year-old man with reactivation of tuberculosis
were noted, including a slightly elevated myeloid-to- from his childhood
erythroid ratio. WBC and RBC morphology appeared d. A 40-year-old man who lost blood during surgery to
normal, however. The Prussian blue stain showed abun- repair a fractured leg
dant stainable iron in the marrow macrophages. The
patients CBC revealed a hemoglobin of 10.8g/dL, but 7. Which of the following individuals is at the greatest risk
RBC indices were within reference ranges. RBC mor for the development of anemia of chronic inflammation?
phology was unremarkable. These findings would be a. A 15-year-old girl with asthma
consistent with: b. A 40-year-old woman with type 2 diabetes mellitus
a. Anemia of chronic inflammation c. A 65-year-old man with hypertension
b. Sideroblastic anemia d. A 30-year-old man with severe rheumatoid arthritis
c. Thalassemia
d. Iron deficiency anemia 8. Which of the following assays can be used cost-effectively
in screening for hereditary hemochromatosis?
3. Predict the iron study results for the patient with Hodgkin a. Polymerase chain reaction
lymphoma described in question 2. b. Ferritin
c. Percent transferrin saturation
Serum % Serum
d. Total bilirubin
Iron Transferrin Ferritin
Level TIBC Saturation Level
a. Decreased Increased Decreased Decreased
b. Increased Normal Increased Normal
c. Increased Increased Normal Increased
d. Decreased Decreased Decreased Increased
266 PART IV Hematopathology: Erythrocyte Disorders

9. In the pathogenesis of the anemia of chronic inflamma- 10. Sideroblastic anemias are anemias that result from:
tion, hepcidin levels: a. Sequestration of iron in hepatocytes
a. Decrease during inflammation and reduce iron b. Inability to incorporate heme into apohemoglobin
absorption from enterocytes c. Sequestration of iron in myeloblasts
b. Increase during inflammation and reduce iron d. Failure to incorporate iron into protoporphyrin IX
absorption from enterocytes
c. Increase during inflammation and increase iron
absorption from enterocytes
d. Decrease during inflammation and increase iron
absorption from enterocytes

REFERENCES
1. Hallberg L: Bioavailability of dietary iron in man. Annu Rev 17. Huebers HA, Beguin Y, Pootrakul P, et al: Intact transferrin
Nutr 1:123-147, 1981. receptors in human plasma and their relation to erythropoi-
2. Suominen P, Punnonen K, Rajamaki A, et al: Serum transfer- esis. Blood 75:102-107, 1990.
rin receptor and transferrin receptorferritin index identify 18. OSullivan DJ, Higgins PG, Wilkinson JF: Oral iron com-
healthy subjects with subclinical iron deficits. Blood 92:2934- pounds: a therapeutic comparison. Lancet 2:482-485, 1955.
2939, 1998. 19. Swan HT, Jowett GH: Treatment of iron deficiency with
3. Andrews N: Disorders of iron metabolism. In Handin RI, Lux ferrous fumarate: assessment by a statistically accurate
SE, Stossel TP, editors: Blood: principles and practice of hemato method. BMJ 2:782-787, 1959.
logy, ed 2, Philadelphia, 2003, Lippincott Williams & Wilkins, 20. Cartwright GE: The anemia of chronic disorders. Semin
pp 1399-1434. Hematol 3:351-375, 1966.
4. Cogswell ME, Looker AC, Pfeiffer CM, et al: Assessment of 21. Sears DA: Anemia of chronic disease. Med Clin North Am
iron deficiency in US preschool children and nonpregnant 76:567-579, 1992.
females of childbearing age: National Health and Nutrition 22. Nemeth E, Tuttle MS, Powelson J, et al: Hepcidin regulates
Examination Survey 2003-2006, Am J Clin Nutr 89:1334- iron efflux by binding to ferroportin and inducing its inter-
1342, 2009. nalization. Science 306:2090-2093, 2004.
5. Green R, Charlton R, Seftel H, et al: Body iron excretion in 23. Rivera S, Liu L, Nemeth E, et al: Hepcidin excess induces the
man: a collaborative study. Am J Med 45:336-353, 1968. sequestration of iron and exacerbates tumor-associated
6. Saarinen UM, Siimes MA, Dallman PR: Iron absorption in anemia. Blood 105:1797-1802, 2005.
infants: high bioavailability of breast milk iron as indicated 24. Nicolas G, Bennoun M, Devaux I, et al: Lack of hepcidin gene
by the extrinsic tag method of iron absorption and by the expression and severe tissue iron overload in upstream stimu-
concentration of serum ferritin. J Pediatr 91:36-39, 1977. latory factor 2 (USF2) knockout mice. Proc Natl Acad Sci
7. Siimes MA, Vuori E, Kuitunen P: Breast milk iron: a declining U S A 98:8780-8785, 2001.
concentration during the course of lactation. Acta Paediatr 25. Nicolas G, Chauvet C, Viatte L, et al: The gene encoding the
Scand 68:29-31, 1979. iron regulatory peptide hepcidin is regulated by anemia,
8. Beutler E, Larsh SE, Gurney CW: Iron therapy in chronically hypoxia, and inflammation. J Clin Invest 110:1037-1044,
fatigued, non-anemic women: a double blind study. Ann 2002.
Intern Med 52:378-394, 1960. 26. Masson PL, Heremans JF, Schonne E: Lactoferrin: an iron
9. Thompson WG, Meola T, Lipkin M Jr, et al: Red cell distribu- binding protein in neutrophilic leukocytes. J Exp Med
tion width, mean corpuscular volume, and transferrin satura- 130:643-658, 1969.
tion in the diagnosis of iron deficiency. Arch Intern Med 27. Ganz T: Hepcidin, a key regulator of iron metabolism and
148:2128-2130, 1988. mediator of anemia in inflammation. Blood 102:783-788,
10. McClure S, Custer E, Bessman JD: Improved detection of early 2003.
iron deficiency in nonanemic subjects. JAMA 253:1021-1023, 28. Means RT Jr, Krantz SB: Progress in understanding the patho-
1985. genesis of the anemia of chronic disease. Blood 80:1639-
11. Charlton RW, Bothwell TH: Definition, prevalence and 1647, 1992.
prevention of iron deficiency. Clin Haematol 11:309-325, 29. Mast AE, Blinder MA, Gronowski AM, et al: Clinical utility of
1982. the soluble transferrin receptor and comparison with serum
12. Cox LA, Adrian GS: Postransciptional regulation of chimeric ferritin in several populations. Clin Chem 44:45-51, 1998.
human transferrin genes by iron. Biochemistry 32:4738-4745, 30. Pincus T, Olsen NJ, Russell IJ, et al: Multicenter study of
1993. recombinant human erythropoietin in correction of anemia
13. Sinniah R, Doggart JR, Neill DW: Diurnal variations of the in rheumatoid arthritis. Am J Med 89:161-168, 1990.
serum iron in normal subjects and in patients with haemo- 31. Arndt U, Kaltwasser JP, Gottschalk R, et al: Correction of iron-
chromatosis. Br J Haematol 17:351-358, 1969. deficient erythropoiesis in the treatment of anemia of chronic
14. Crosby WH, ONeil-Cutting MA: A small-dose iron tolerance disease with recombinant human erythropoietin. Ann
test as an indicator of mild iron deficiency. Clin Invest Hematol 84:159-166, 2005.
251:1986-1987, 1984. 32. Horrigan DL, Harris JW: Pyridoxine responsive anemia: anal-
15. Brugnara C, Schiller B, Moran J: Reticulocyte hemoglobin ysis of 62 cases. Adv Intern Med 12:103-174, 1964.
equivalent (Ret He) and assessment of iron deficient states. 33. Verwilghen R, Reybrouck G, Callens L, et al: Antituberculosis
Clin Lab Haem 29:303-306, 2006. drugs and sideroblastic anemia. Br J Haematol 11:92-98,
16. Lamola AA, Joselow M, Yamane T: Zinc protoporphyrin 1965.
(ZPP): a simple, sensitive fluorometric screening test for lead 34. Benson P: Lead poisoning in children. Dev Med Child Neurol
poisoning. Clin Chem 21:93-97, 1975. 7:569-571, 1965.
CHAPTER 19 Disorders of Iron and Heme Metabolism 267
35. Campbell AM, Williams ER: Chronic lead intoxication mim- 44. Lebron JA, Bjorkman PJ: The transferrin receptor binding
icking motor neurone disease. BMJ 4:582, 1968. site on HFE, the class I MHC-related protein mutated in
36. Granick S, Sassa S, Granick JL, et al: Assays for porphyrins, hereditary hemochromatosis. J Mol Biol 289:1109-1118,
delta-aminolevulinic acid dehydratase, and porphyrinogen 1999.
synthetase in microliter samples of blood: application to 45. Andrews NC, Levy JE: Iron is hot: an update on the
metabolic defects involving the heme pathway. Proc Natl pathophysiology of hemochromatosis. Blood 92:1845-1851,
Acad Sci U S A 69:2381-2385, 1972. 1998.
37. Lachant NA, Tomoda A, Tanaka KR: Inhibition of the pentose 46. Pietrangelo A: Herediary hemochromatosisa new look at
phosphate shunt by lead: a potential mechanism for hemo- an old disease. N Engl J Med 350:2383-2398, 2004.
lysis in lead poisoning. Blood 63:518-524, 1984. 47. McCord JM: Iron, free radicals, and oxidative injury. Semin
38. Sassa S, Shibahara S: Disorders of heme production and Hematol 35:5-12, 1998.
catabolism. In Handin RI, Lux SE, Stossel TP, editors: Blood: 48. Vautier G, Bomford AB, Portmann BC, et al: p53 mutations
principles and practice of hematology, Philadelphia, 2003, in British patients with hepatocellular carcinoma: clustering
Lippincott Williams & Wilkins, pp 1435-1502. in genetic hemochromatosis. Gastroenterology 117:154-160,
39. Valentine WN, Paglia DE, Fink K, et al: Lead poisoning. Associa- 1999.
tion with hemolytic anemia, basophilic stippling, erythrocyte 49. Hussain SP, Raja K, Amstad PA, et al: Increased p53 mutation
pyrimidine 5-nucleotidase deficiency, and intraerythrocytic load in nontumorous human liver of Wilson disease and
accumulation of pyrimidines. J Clin Invest 58:926-932, 1976. hemochromatosis: oxyradical overload diseases. Proc Natl
40. Edwards CQ, Griffen LM, Ajioka RS, et al: Screening Acad Sci U S A 97:12770-12775, 2000.
for hemochromatosis: phenotype versus genotype. Semin 50. Niederau C, Strohmeyer G, Stremmel W: Long term
Hematol 35:72-76, 1998. survival in patients with hereditary hemochromatosis. Gastro
41. Feder JN, Gnirke A, Thomas W, et al: A novel MHC class I-like enterology 110:1107-1119, 1996.
gene is mutated in patients with hereditary haemochromato- 51. Bothwell TH, MacPhail AP: Hereditary hemochromatosis:
sis. Nat Genet 13:399-408, 1996. etiologic, pathologic, and clinical aspects. Semin Hematol
42. Waheed A, Parkkila S, Zhou XY, et al: Hereditary hemochro- 35:55-71, 1998.
matosis: effect of C282Y and H63D mutations on association 52. Phatak PD, Guzman G, Woll JE, et al: Cost effectiveness of
with 2-microglobulin, intracellular processing, and cell screening for hemochromatosis. Arch Intern Med 154:769-
surface expression of the HFE protein in COS-7 cells. Proc Natl 776, 1994.
Acad Sci U S A 94:12384-12389, 1997. 53. Cappellini MD, Pattoneri P. Oral iron chelators. Annu Rev
43. Salter-Cid L, Brunmark A, Li Y, et al: Transferrin receptor is Med 60:25-38, 2009.
negatively modulated by the hemochromatosis protein HFE: 54. Imran F, Phatak P. Pharmacoeconomic benefits of deferasirox
implications for cellular iron homeostasis. Proc Natl Acad Sci in the management of iron overload syndromes. Expert Rev
U S A 96:5435-5439, 1999. Pharmacoecon Outcomes Res 9:297-304, 2009.
20 Anemias Caused by Defects of
DNA Metabolism
Linda H. Goossen*

OUTLINE OBJECTIVES
Etiology After completion of this chapter, the reader will be able to:
Physiologic Roles of
Vitamin B12 and Folate 1. Discuss the relationships among macrocytic anemia, stance such as pregnancy, drug regimens, or patho-
Defect in Megaloblastic megaloblastic anemia, and pernicious anemia, logic conditions.
Anemia due to Deficiency and classify anemias appropriately within these 6. Recognize complete blood count, reticulocyte count,
in Folate and Vitamin B12 categories. red and white blood cell morphologies, and bone
Other Causes of 2. Discuss the physiologic roles of folate and vitamin B12 marrow findings consistent with megaloblastic
Megaloblastosis in DNA production and the general metabolic path- anemia.
Systemic Manifestations ways in which they act. 7. Given the results of tests measuring levels of serum
of Folate and Vitamin 3. Describe the absorption and distribution of vitamin vitamin B12, serum methylmalonic acid, serum folate,
B12 Deficiency B12, including carrier proteins and the biologic activity plasma or serum homocysteine, and antibodies to
Causes of Vitamin
of various vitamin-carrier complexes. intrinsic factor and parietal cells, determine the likely
Deficiencies
4. Describe the biochemical basis for development cause of a patients deficiency.
Folate Deficiency
Vitamin B12 Deficiency of anemia with deficiencies of vitamin B12 and 8. Recognize results of bilirubin and lactate dehydroge-
Laboratory Diagnosis folate, and explain the cause of the accompanying nase tests that are consistent with megaloblastic
Screening Tests megaloblastosis. anemia and explain why the test values are elevated
Specific Diagnostic Tests 5. Recognize individuals at risk for megaloblastic anemia in this condition.
Macrocytic by virtue of age, dietary habits, physiologic circum-
Nonmegaloblastic
Anemias
Treatment

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

During a holiday visit, the children of a 76-year-old man Patient Value Reference Range
noticed that he seemed more forgetful than usual and that he Hct (%) 27 39-55
had difficulty walking. Concerned about the possibility of a MCV (fL) 121.6 80-100
mild stroke, the children insisted that he see his physician. MCH (pg) 38.3 25.4-34.6
The physician diagnosed a peripheral neuropathy affecting MCHC (g/dL) 31.5 31-37
the fathers ability to walk. In addition, the physician noted RDW (%) 18 11.5-13.5
that he was quite pale and slightly jaundiced and ordered Platelets ( 109/L) 115 150-450
routine hematologic studies. The results were as follows: Reticulocytes (%) 1.8 0.5-1.5
WBC differential: unremarkable with the exception of hypersegmentation
Patient Value Reference Range of neutrophils
9
WBCs ( 10 /L) 3.2 4.5-11.0 RBC morphology: moderate anisocytosis, moderate poikilocytosis,
RBCs ( 1012/L) 2.22 4.30-5.90 macrocytes, oval macrocytes, few teardrop cells
Hb (g/dL) 8.5 13.9-16.3 Continued

*The author acknowledges the fine work of Dr. Kathryn Doig, the author of this chapter in the previous edition.

268
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 269

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

Because of these findings, additional tests were ordered, with 2. Is the patients reticulocyte response adequate to compen-
the following results: sate for the anemia?
Serum vitamin B12: decreased 3. Based on the available test results, what can you conclude
Serum folate: within reference range about the cause of the patients anemia?
Serum methylmalonic acid: increased 4. What additional testing would be helpful to diagnose the
1. Which of the CBC findings led the physician to order the specific cause of this patients anemia?
vitamin assays?

I
mpaired deoxyribonucleic acid (DNA) metabolism causes
systemic effects by impairing production of all rapidly
BOX 20-1 Classification of Macrocytic Anemias by
dividing cells of the body. These are chiefly the cells of
Cause
the skin, the epithelium of the gastrointestinal tract, and the
Megaloblastic anemia
hematopoietic tissues. Because these all must be replenished
Folate deficiency
throughout life, any impairment of cell production is evident
Inadequate intake
in these tissues first. Patients may experience symptoms in any
Increased need
of these systems, but the blood provides a ready tissue for
Impaired absorption (e.g., inflammatory bowel disease)
analysis. The hematologic effects, especially megaloblastic
Impaired use due to drugs
anemia, have come to be recognized as the hallmark of the
Excessive loss with renal dialysis
diseases affecting DNA metabolism.
Vitamin B12 deficiency
Inadequate intake
ETIOLOGY Increased need
Impaired absorption
The root cause of megaloblastic anemia is impaired DNA syn-
Failure to split from food
thesis. The anemia is named for the very large cells of the bone
Failure to split from haptocorrin
marrow that develop a distinctive morphology (see section on
Lack of intrinsic factor
laboratory diagnosis) due to a reduction in the number of cell
Pernicious anemia
divisions. Megaloblastic anemia is one example of a macrocytic
Helicobacter pylori infection
anemia. Box 20-1 shows the classification of macrocytic
Gastrectomy
anemias. Understanding the etiology of megaloblastic anemia
Hereditary intrinsic factor deficiency
requires a review of DNA synthesis with particular attention to
General malabsorption (e.g., inflammatory bowel disease)
the roles of vitamin B12 (cobalamin) and folic acid (folate).
Inherited errors in absorption or transport
Imerslund-Grsbeck syndrome
Physiologic Roles of Vitamin B12 and Folate Transcobalamin deficiency
Vitamin B12 (cobalamin) is an essential nutrient consisting of
Competition for the vitamin
a tetrapyrrole (corrin) ring containing cobalt that is attached
Diphyllobothrium latum (fish tapeworm) infection
to 5,6-dimethylbenzimidazolyl ribonucleotide (Figure 20-1).
Blind loop syndrome
Vitamin B12 is a coenzyme in two biochemical reactions in
Other causes of megaloblastosis
humans. One is isomerization of methylmalonyl coenzyme A
Myelodysplastic syndrome
(CoA) to succinyl CoA, which requires vitamin B12 (in the
Acute erythroleukemia (FAB-M6)
adenosylcobalamin form) as a cofactor and is catalyzed by
Congenital dyserythropoietic anemia
the enzyme methylmalonyl CoA mutase (Figure 20-2). In
Reverse transcriptase inhibitors
the absence of vitamin B12, the impaired activity of methyl-
Macrocytic, nonmegaloblastic anemias
malonyl CoA mutase leads to a high level of serum methylma-
Normal newborn status
lonic acid, which is useful for the diagnosis of vitamin B12
Reticulocytosis
deficiency (discussed in the section on laboratory diagnosis).
Liver disease
The second reaction is the transfer of a methyl group from
Chronic alcoholism
5-methyltetrahydrofolate (5-methyl THF) to homocysteine,
Bone marrow failure
which thereby generates methionine. This reaction is catalyzed
270 PART IV Hematopathology: Erythrocyte Disorders

X by the enzyme methionine synthase and uses vitamin B12 (in


the methylcobalamin form) as a coenzyme (discussed later in
R R this section). Methylcobalamin is synthesized through reduc-
tion and methylation of vitamin B12. This reaction represents
B the link between folate and vitamin B12 coenzymes and appears
R N to account for the requirement for both vitamins in normal
erythropoiesis.1,2
R A N Co N C Folate is the general term used for any form of the vitamin
folic acid. Folic acid is the synthetic form in supplements and
N R
H fortified food. Folates consist of a pteridine ring attached to
D para-aminobenzoate with one or more glutamate residues
(Figure 20-3). The function of folate is to transfer carbon units
R
O in the form of methyl groups from donors to receptors. In this
N capacity, folate plays an important role in the metabolism of
NH amino acids and nucleotides. Deficiency of the vitamin leads
OH to impaired cell replication and other metabolic alterations.
O
N Folate circulates in the blood predominantly as 5-methyl THF.3
O
P

O N N
O NH2 C C CH
OH N C C CH2 N CO glutamate
C N
H
OH: hydroxycobalamin OH
CH3: methylcobalamin Folic acid
X Ado: 5-deoxyadenosylcobalamin
CN: cyanocobalamin
N N H
NH2 C C C
Figure 20-1 Structure of vitamin B12 (cobalamin) and its analogs, H
hydroxycobalamin and cyanocobalamin (forms often found in food and sup- N C CH CH2 N CO glutamate
plements) and methylcobalamin and 5-deoxyadenosylcobalamin (coenzyme C N
H H
forms). The basic structure of cobalamin includes a tetrapyrrole (corrin) ring OH
with a central cobalt atom linked to 5,6-dimethylbenzimidazolyl ribonucleo- Tetrahydrofolate
tide. (From Scott JM, Browne P: Megaloblastic anemia. In Caballero B,
Allen L, Prentice A, editors: Encyclopedia of human nutrition, ed 2, Oxford, H
2006, Academic Press, p 113.) N N H
NH2 C C C
H
N C CH CH2 N CO glutamate
C N
H CHO
OH
10-Formyltetrahydrofolate

COOH H
N H N
HCCH3 NH2 C C C
H
CSCoA N C CH CH2 N CO glutamate
C N CH2
O Methylmalonyl CoA OH
Methylmalonyl CoA mutase Vitamin B12 5,10-Methylenetetrahydrofolate

COOH H
N N H
CH2 NH2 C C C
H
N C CH CH2 N CO glutamate
CH2
C N
H
CSCoA OH CH3
O Succinyl CoA 5-Methyltetrahydrofolate
Figure 20-2 Role of vitamin B12 in the metabolism of methylmalonyl Figure 20-3 Structure of synthetic folic acid and the naturally occurring
coenzyme A (CoA). Vitamin B12, in the 5-deoxyadenosylcobalamin form, is forms of the vitamin. (From Scott JM, Browne P: Megaloblastic anemia. In
a coenzyme in the isomerization of methylmalonyl CoA to succinyl CoA. The Caballero B, Allen L, Prentice A, editors: Encyclopedia of human nutrition,
reaction is catalyzed by the enzyme methylmalonyl CoA mutase. ed 2, Oxford, 2006, Academic Press, p 114.)
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 271

Uridine Thymidine dATP dCTP dGTP

3
dUMP dTMP dTTP DNA

5,10-methylene THF DHF

Glycine
2 4
Serine THF*
*Polyglutamation of THF (addition of 1-6 more glutamic
acid residues) is essential for its retention in cell
Methionine

Vitamin B12 1 Site of


folate trap
Homocysteine
5-methyl THF 1 Methionine synthase and vitamin
B12 (in methylcobalamin form)
Cytoplasm
2 Serine hydroxymethyl transferase
Plasma and vitamin B6
5-methyl THF 3 Thymidylate synthetase
4 Dihydrofolate reductase

Figure 20-4 Role of folate and vitamin B12 in DNA synthesis. Folate enters the cell as 5-methyltetrahydrofolate (5-methyl THF). In the cell, a methyl group
is transferred from 5-methyl THF to homocysteine, converting it to methionine and generating tetrahydrofolate (THF). This reaction is catalyzed by methionine
synthase and requires vitamin B12 as a cofactor. THF is then converted to 5,10-methylene THF by the donation of a methyl group from serine. The methyl
group of 5,10-methylene THF is then transferred to deoxyuridine monophosphate (dUMP), which converts it to deoxythymidine monophosphate (dTMP) and
converts 5,10-methylene THF to dihydrofolate (DHF). This reaction is catalyzed by thymidylate synthetase. dTMP is a precursor of deoxythymidine triphosphate
(dTTP), which is used to synthesize DNA. THF is regenerated by the conversion of DHF to THF by the enzyme DHF reductase. A deficiency of vitamin B12
prevents the production of THF from 5-methyl THF; as a result, folate becomes metabolically trapped as 5-methyl THF. This constitutes the folate trap.

5-Methyl THF is metabolically inactive until it is demethylated Defect in Megaloblastic Anemia due to
to tetrahydrofolate (THF), whereupon folate-dependent reac- Deficiency in Folate and Vitamin B12
tions may take place. When either folate or vitamin B12 is missing, thymidine nucleo-
Folate has an important role in DNA synthesis. As seen in tide production for DNA synthesis is impaired. Folate defi-
Figure 20-4, within the cytoplasm of the cell, a methyl group ciency has the more direct effect, ultimately preventing the
is transferred from 5-methyl THF to homocysteine, which methylation of dUMP. The effect of vitamin B12 deficiency is
converts it to methionine and generates THF. This reaction is more indirect, preventing the production of THF from 5-methyl
catalyzed by the enzyme methionine synthase and requires THF. When vitamin B12 is deficient, progressively more and
vitamin B12 in the form of methylcobalamin as a cofactor. more of the folate becomes metabolically trapped as 5-methyl
THF is then converted to 5,10-methylenetetrahydrofolate THF. This constitutes what has been called the folate trap as
(5,10-methylene THF); the methyl group for this reaction 5-methyl THF accumulates and is unable to supply the folate
comes from serine as it is converted to glycine. The methyl cycle with THF. Some 5-methyl THF also leaks out of the cell
group of 5,10-methylene THF is then transferred to deoxyuri- if it is not readily polyglutamated. This results in a decrease in
dine monophosphate (dUMP), which converts it to deoxythy- intracellular folate.5 In addition, when either folate or vitamin
midine monophosphate (dTMP). This reaction is catalyzed B12 is deficient, homocysteine accumulates, because vitamin B12
by thymidylate synthetase and results in the conversion of is unable to convert it to methionine (see Figure 20-4).
5,10-methylene THF to dihydrofolate (DHF). Deoxythymidine In this state of diminished thymidine availability, uridine is
monophosphate is a precursor to deoxythymidine triphos- incorporated into DNA.6,7 The DNA repair process can remove
phate (dTTP), which, like the other nucleotide triphosphates, the uridine, but without available thymidine, the repair process
is a building block of the DNA molecule. THF is regenerated is unsuccessful. Although the DNA can unwind and replication
by the conversion of DHF to THF by the enzyme DHF reduc- can begin, at any point where a thymidine nucleotide is needed,
tase. Because some of the folate is catabolized during the cycle, there is essentially an empty space in the replicated DNA
the regeneration of THF also requires additional 5-methyl THF sequence, which results in many single-strand breaks. When
from the plasma. It is important to note that once in the cell, excisions at opposing DNA strand sites coincide, double-strand
folate is rapidly polyglutamated by the addition of one to six breaks occur. Repeated DNA strand breaks lead to fragmenta-
glutamic acid residues. This conjugation is required for reten- tion of the DNA strand.4 The resulting DNA is nonfunctional,
tion of THF in the cell and it also promotes attachment of and the DNA replication process is incomplete. Cell division is
folate to enzymes.4 halted, which results in either cell lysis or apoptosis8 of many
272 PART IV Hematopathology: Erythrocyte Disorders

megaloblastic RBCs seen in the vitamin deficiencies. In addi-


BOX 20-2 Disruption of the Folate Cycle in Cancer tion, nuclear-cytoplasmic asynchrony and megaloblastic RBCs
Chemotherapy may be seen in congenital dyserythropoietic anemia (CDA)
The central role of folate in cell division makes it a target for che- types I and III (see Chapter 21). The CDAs are rare conditions
motherapeutic drugs used to treat cancer. Folate analogues can be that usually manifest in childhood and may be distinguished
used to compete for folate in DNA production and result in impaired from the acquired causes of megaloblastosis by clinical history
cell division. The cells in the cell cycle, such as cancer cells and and morphologic differences. In CDA I internuclear chromatin
other normally rapidly dividing cells such as epithelium and blood bridging of erythroid cells or binucleated forms are observed,
cells, are most susceptible to the drug interference. Methotrexate, and in CDA III giant multinucleated erythroblasts are present.
used in the treatment of leukemia and arthritis, is an example of a Another rare condition in which RBC precursors have a mega-
folate antimetabolite drug. It has a higher affinity for dihydrofolate loblastic appearance is acute erythroleukemia, previously clas-
reductase than does tetrahydrofolate. Thus, methotrexate enters the sified as FAB M6 (see Chapter 36). In this condition, the cells
folate cycle in preference to tetrahydrofolate, and the folate cycle is are macrocytic, and the immature appearance of the nuclear
blocked by the drug. Methotrexate treatment typically is followed by chromatin is similar to the more open appearance of the chro-
what is known as leucovorin rescue. Leucovorin is a folic acid deriva- matin in megaloblasts. There are usually other aberrant find-
tive that can be administered to counteract the effects of methotrex- ings in erythroleukemia, including an increase of myeloblasts
ate or other folate antagonists. in the bone marrow; however, an experienced morphologist
can discern the subtle differences. Reverse transcriptase inhibi-
tors, used to treat human immunodeficiency virus (HIV) infec-
erythroid cells. Cells that do not lyse are released into the cir- tions, interfere with DNA production and may also lead to
culation. The dependency of DNA production on folates has megaloblastic changes.10
been used in cancer chemotherapy (Box 20-2). Although the conditions described in this section are char-
In the case of red blood cell (RBC) development, these acterized by megaloblastic morphology, they are due to
vitamin deficiencies result in ineffective hematopoiesis with acquired or inherited mutations in progenitor cells or interfer-
increased apoptosis of erythroid progenitor cells. The remain- ence with DNA synthesis and are refractive to therapy with
ing erythroid cells are larger than those cells that are normally vitamin B12 or folic acid.
seen during the final stages of erythropoiesis and their nuclei
are immature-appearing compared with the cytoplasm (asyn-
SYSTEMIC MANIFESTATIONS OF FOLATE
chrony). In contrast to the normally dense chromatin of com-
AND VITAMIN B12 DEFICIENCY
parable normoblasts, megaloblastic erythroid precursors have
an open, finely stippled, reticular pattern.5 The nuclear changes When DNA synthesis and subsequent cell division are impaired
seen in the megaloblastic cells are related to cell cycle delay, by lack of folate or vitamin B12, megaloblastic anemia and its
prolonged resting phase, and arrest in nuclear maturation. systemic manifestations develop. With either vitamin defi-
Electron microscopy has revealed that reduced synthesis of ciency, patients may experience general symptoms related to
histones is also responsible for morphologic changes in the the anemia (fatigue, weakness, and shortness of breath) and
chromatin of megaloblastic cells, regardless of the causes.9 symptoms related to the alimentary tract. The loss of epithe-
Because ribonucleic acid (RNA) contains uracil instead of thy- lium on the tongue results in a smooth surface and soreness
midine residues, RNA function is not affected by vitamin B12 (glossitis). Loss of epithelium along the gastrointestinal tract
or folate deficiency. Thus cytoplasmic development is not gen- can result in gastritis, nausea, or constipation.
erally disturbed in these cells, which results in the asynchrony Although the blood pictures seen with the two vitamin
between cytoplasm and nucleus. Together, the accumulation of deficiencies are indistinguishable, the clinical presentations
cells at earlier stages of differentiation and cells with increased vary. In vitamin B12 deficiency, neurologic symptoms may be
size and immaturity result in the appearance of erythroid cells pronounced and may even occur in the absence of anemia.5
in the bone marrow that are pathognomonic of megaloblastic These include memory loss, numbness and tingling in toes and
anemia.8 Due to the ineffective hematopoiesis, pancytopenia fingers, loss of balance, and further impairment of walking
is also evident, with certain distinctive cellular changes (see by loss of vibratory sense, especially in the lower limbs.11
section on laboratory diagnosis). Neuropsychiatric symptoms may also be present, including
personality changes and psychosis. These symptoms seem to
Other Causes of Megaloblastosis be the result of demyelinization of the spinal cord and peri
Vitamin B12 and folate deficiency are not the only causes pheral nerves, but the relationship of this demyelinization to
of megaloblastic erythrocytes. Dysplastic erythroid cells in vitamin B12 deficiency is unclear. The role of increases in tumor
myelodysplastic syndrome (MDS) can also have megaloblas- necrosis factor-, a neurotoxic agent, and decreases in epider-
toid features (see Chapter 35). In MDS, the macrocytic eryth- mal growth factor, a neurotrophic agent, in the development
rocytes and their progenitors characteristically show delayed of neurologic symptoms in vitamin B12-deficient patients is
cytoplasmic and nuclear maturation, including cytoplasmic being researched.12,13
vacuole formation, nuclear budding, multinucleation, and At one time, folate deficiency was believed to be more
nuclear fragmentation, and thus may be distinguished from the benign clinically than vitamin B12 deficiency. Later research
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 273

suggested that low levels of folate and the resulting high homo-
cysteine levels were risk factors for cardiovascular disease.14 BOX 20-3 Some Drugs That May Lead to Impaired
More recent research has not substantiated this association,15-17 Use of Folate
although some point out that high folate levels provide a
cardioprotective effect in diabetic patients and certain ethnic Impair Folate Metabolism
populations.18,19 The evidence at this time is unclear as to Methotrexate (Trexall): antiarthritic, chemotherapeutic
whether persistent suboptimal folate status may have a signifi- 5-Fluorouracil (Adrucil): chemotherapeutic
cant long-term health impact. In addition, there is evidence of Hydroxyurea (Hydrea): antimetabolite
depression, peripheral neuropathy, and psychosis related to Pyrimethamine (Daraprim): antibacterial
folate deficiency.20-22 Folate levels appear to influence the effec- Pentamidine (Pentam): antimicrobial
tiveness of treatments for depression.23 Folate deficiency during Phenytoin (Dilantin): anticonvulsant
pregnancy can result in impaired formation of the fetal nervous
system, resulting in neural tube defects such as spina bifida,24 Impair Folate Absorption
despite the fact that the fetus accumulates folate at the expense Metformin (Glucophage): oral antidiabetic
of the mother. Pregnancy requires a considerable increase in Cholestyramine (Questran): cholesterol lowering
folate to fulfill the requirements related to rapid fetal growth,
uterine expansion, placental maturation, and expanded blood
volume.3 Ensuring adequate folate levels in women of child- Folate absorption may also be impaired by intestinal
bearing age is particularly important because many women are disease, especially sprue. Sprue is characterized by weakness,
likely to be unaware of their pregnancy during the first crucial weight loss, and steatorrhea (fat in the feces), which is evidence
weeks of fetal development. Fortification of the U.S. food that the intestine is not absorbing food properly. It is seen in
supply with folic acid in grain and cereal products was man- the tropics (tropical sprue), where its cause is generally consid-
dated by the Food and Drug Administration in 1998 to lower ered to be overgrowth of enteric pathogens.29 Celiac disease
the risk of neural tube defects in the unborn. (nontropical sprue) has been traced to intolerance of the gluten
in some grains29 (gluten-induced enteropathy) and can be con-
trolled by eliminating wheat, barley, and rye products from the
CAUSES OF VITAMIN DEFICIENCIES
diet. Surgical resection of the small intestine and inflammatory
In general, vitamin deficiencies may arise because the vitamin bowel disease can also decrease folate absorption.
is in relatively short supply, because use of the vitamin is
impaired, or because excessive loss is occurring. Folate defi- Impaired Use of Folate
ciency can be caused by all of these mechanisms. Numerous drugs impair folate metabolism (Box 20-3).30,31
Antiepileptic drugs are particularly known for this,32 and the
Folate Deficiency result is macrocytosis with frank megaloblastic anemia. In
Inadequate Intake most instances, folic acid supplementation is sufficient to over-
The most common cause of folate deficiency is inadequate ride the impairment and allow the patient to continue therapy.33
dietary intake.25 Folate is ubiquitous in foods, but a generally
poor diet can result in deficiency. Good sources of folate Excessive Loss of Folate
include leafy green vegetables, dried beans, liver, beef, fortified Physiologic loss of folate occurs through the kidney. The
breakfast cereals, and some fruits, especially oranges.26 Folates amount is small and not a cause of deficiency. Patients under-
are heat labile, and overcooking of foods can diminish their going renal dialysis lose folate in the dialysate, however; thus
nutritional value.3 supplemental folic acid is routinely provided to these individu-
als to prevent megaloblastic anemia.11
Increased Need
Increased need for folate occurs during pregnancy and lactation Vitamin B12 Deficiency
when the mother must supply her own needs plus those of the Inadequate Intake
fetus or infant. Infants and children also have increased need True dietary deficiency of vitamin B12 is possible for strict veg-
for folate during growth.3 etarians (vegans) who do not eat meat, eggs, or dairy products.
Although it is an essential vitamin for animals, plants cannot
Impaired Absorption synthesize vitamin B12, and it is not available from vegetable
Folates are absorbed from the diet in the small intestine; sources. The best dietary sources are animal products such as
however, only 50% of what is ingested is available for absorp- liver, milk, cheese, and eggs.25
tion.3 A rare autosomal recessive deficiency of a folate trans-
porter protein (PCFT/HCP1) severely decreases intestinal Increased Need
absorption of folate.27 Once across the intestinal cell, most Increased need for vitamin B12 occurs during pregnancy, lacta-
folate is transported in the plasma as 5-methyl THF unbound tion, and growth. Due to the vigorous cell replication, what
to any specific carrier.11 Its entry into cells, however, is carrier would otherwise be a diet adequate in vitamin B12 can become
mediated.28 inadequate during these periods.
274 PART IV Hematopathology: Erythrocyte Disorders

Impaired Absorption the vitamin B12intrinsic factor compound, and megalin, a


Vitamin B12 in food is released from food proteins primarily in membrane transport protein.34-37 Once in the enterocyte, the
the acid environment of the stomach, aided by pepsin, and is vitamin B12 is then freed from intrinsic factor and bound to
subsequently bound by a specific salivary protein, haptocorrin, transcobalamin (previously called transcobalamin II) and
also known as R-binder protein (Figure 20-5). In the small intes- released into the circulation. In the plasma, only 20% of the
tine, vitamin B12 is released from haptocorrin by the action of vitamin B12 is bound to transcobalamin; the remaining 80% is
pancreatic proteases, including trypsin. It is then bound by bound to transcobalamin I and III, referred to as the haptocor-
intrinsic factor, produced by the gastric parietal cells. Vitamin rins.34,38 The vitamin B12transcobalamin complex, termed holo-
B12 binding to intrinsic factor is required for absorption by the transcobalamin, is the metabolically active form of vitamin B12.
ileal cells (enterocytes) that possess receptors for the complex. Holotranscobalamin binds to specific receptors on the surfaces
These receptors are cubilin-amnionless complex, which binds of many different types of cells and enters the cells by

CBL P

Pepsin
HCI

P
Pancreatic
proteases CBL
R
Stomach
R CBL Saliva

IF IF IF
Duodenum
CBL

CBL IF Cubilin/
IF amnionless/
CBL megalin

CBL Enterocyte
IF
Ileum
CBL TC TC

Portal
circulation
CBL TC

Biliary
excretion
CBL CBL CBL CBL
TC TC
Methyl-
Storage CBL
TC-R TC-R
TC TC

Hepatocyte Pronormoblast
Figure 20-5 Normal absorption of vitamin B12. Dietary vitamin B12 (cobalamin, CBL) is protein (P) bound. In the stomach, pepsin and hydrochloric acid
(HCl) secreted by parietal cells release CBL from P. CBL then binds salivary haptocorrin (R-binder protein, R) and remains bound until intestinal pancreatic
proteases, including trypsin, catalyze its release. Parietal cells also secrete intrinsic factor (IF), which binds CBL in the duodenum. Cubilin-amnionless and
megalin receptors in ileal enterocytes bind CBL-IF and release the CBL. Enterocytes produce transcobalamin (TC), which binds CBL and transports it through
the portal circulation. Bone marrow pronormoblast membrane TC receptors (TC-R) bind CBL-TC and release the CBL, which is converted to methylcobalamin
(methyl-CBL). Methyl-CBL is a coenzyme that supports homocysteine-methionine conversion. Hepatocyte TC-R receptors bind CBL-TC and release the CBL,
which is moved to storage organelles or excreted through the biliary system.
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 275
endocytosis, with subsequent release of vitamin B12 from the collection and the use of radioactive cobalt in vitamin B12 to
carrier.39 The body maintains a substantial reserve of absorbed trace absorption, it has fallen from favor, and safer diagnostic
vitamin B12 in hepatocytes. tests are preferred (see section on laboratory diagnosis).
The absorption of vitamin B12 can be impaired by (1) failure Another feature of the autoimmune response in pernicious
to separate vitamin B12 from food proteins in the stomach, anemia is the production of antibodies to intrinsic factor43 and
(2) failure to separate vitamin B12 from haptocorrin in the gastric parietal cells44 that are detectable in serum. The most
intestine, (3) lack of intrinsic factor, (4) malabsorption, and common antibody to intrinsic factor blocks the site on intrinsic
(5) competition for available vitamin. factor where vitamin B12 binds,41 which inhibits the formation
of the intrinsic factorvitamin B12 complex and prevents the
Failure to Separate Vitamin B12 from Food Proteins. A absorption of the vitamin. These blocking antibodies are
condition known as food-cobalamin malabsorption is character- present in about 70% of patients with pernicious anemia.41
ized by hypochlorhydria and the resulting inability of the body Parietal cell antibodies are detectable in about 90% of patients
to release vitamin B12 from food or intestinal transport proteins with pernicious anemia.41
for subsequent binding to intrinsic factor. Food-cobalamin Other causes of lack of intrinsic factor. A lack of
malabsorption is caused primarily by atrophic gastritis or intrinsic factor may also be related to H. pylori infection. Left
atrophy of the stomach lining that often occurs with increasing untreated, colonization of the gastric mucosa with H. pylori
age.40 Because histamine 2 receptor blockers and proton pump progresses until the parietal cells are entirely destroyed, a
inhibitors lower gastric acidity, the long-term use of these drugs process involving both local and systemic immune pro-
for the treatment of ulcers and gastroesophageal reflux disease, cesses.45,46 In addition, partial or total gastrectomy, which result
as well as gastric bypass surgery, also induce food-cobalamin in removal of intrinsic factorproducing parietal cells, invari-
malabsorption.40 ably leads to vitamin B12 deficiency.
Impaired absorption of vitamin B12 can also be caused by
Failure to Separate Vitamin B12 from Haptocorrin. Lack hereditary intrinsic factor deficiency. This is a rare autosomal
of gastric acidity or lack of trypsin as a result of chronic pan- recessive disorder characterized by the absence or nonfunction-
creatic disease can prevent vitamin B12 absorption because the ality of intrinsic factor. In contrast to the acquired forms of
vitamin remains bound to haptocorrin in the intestine and pernicious anemia, histology and gastric acidity are normal.36
unavailable to intrinsic factor.11
Malabsorption. General malabsorption of vitamin B12
Lack of Intrinsic Factor. Lack of intrinsic factor consti- can be caused by the same conditions interfering with folate
tutes a significant cause of impaired vitamin B12 absorption. It absorption, such as celiac disease, tropical sprue, and inflam-
is most commonly due to autoimmune disease, as in perni- matory bowel disease.
cious anemia, but can also result from the loss of parietal cells
with Helicobacter pylori infection, total or partial gastrectomy, Inherited Errors of Vitamin B12 Absorption and Transport.
or hereditary intrinsic factor deficiency. Imerslund-Grsbeck syndrome is a rare autosomal recessive
Pernicious anemia. Pernicious anemia is an autoim- condition caused by mutations in the genes for either cubilin
mune disorder characterized by impaired absorption of vitamin or amnionless. This defect results in decreased endocytosis of
B12 due to a lack of intrinsic factor.41 This condition is called the intrinsic factorvitamin B12 complex by ileal cells. Trans
pernicious anemia because the disease was fatal before its cause cobalamin deficiency is another rare autosomal recessive
was discovered. The incidence is approximately 25 per 100,000 condition resulting in a deficiency of physiologically available
persons older than 40 years of age.5 Pernicious anemia most vitamin B12.36,47
often manifests in the sixth decade or later, but can also be
found in children. Competition for Vitamin B12 . Competition for available
In pernicious anemia, autoimmune lymphocyte-mediated vitamin B12 in the intestine may come from intestinal organ-
destruction of gastric parietal cells severely reduces the amount isms. The fish tapeworm Diphyllobothrium latum is able to split
of intrinsic factor secreted in the stomach. Pathologic CD4 T vitamin B12 from intrinsic factor,48 rendering the vitamin
cells inappropriately recognize and initiate an autoimmune unavailable for host absorption. Also, blind loops, portions of
response against the H+/K+adenosine triphosphatase embed- the intestines that are stenotic as a result of surgery or inflam-
ded in the membrane of the parietal cells.42 A chronic inflam- mation, can become overgrown with intestinal bacteria that
matory infiltration follows, which extends into the wall of the compete effectively with the host for available vitamin B12.11 In
stomach.41 Over a period of years and even decades, there is both of these cases, the host is unable to absorb sufficient
progressive development of atrophic gastritis resulting in the vitamin B12, and megaloblastic anemia results.
loss of the parietal cells with their secretory products, H+ and
intrinsic factor. The loss of H+ production in the stomach con-
LABORATORY DIAGNOSIS
stitutes achlorhydria. Low gastric acidity was previously an
important diagnostic criterion for pernicious anemia. The The tests used in the diagnosis of megaloblastic anemia include
absence of intrinsic factor can also be detected using the Schil- screening tests and specific diagnostic tests to identify the spe-
ling test. However, because the test requires a 24-hour urine cific vitamin deficiency and perhaps its cause.
276 PART IV Hematopathology: Erythrocyte Disorders

Screening Tests of the disease50 and may persist for up to 2 weeks after treat-
Five tests used to screen for megaloblastic anemia are the com- ment is initiated.5 Hypersegmented neutrophils noted in the
plete blood count (CBC), white blood cell (WBC) manual WBC differential report is a significant finding and requires a
differential, reticulocyte count, serum bilirubin, and lactate reporting rule that can be applied consistently, because even
dehydrogenase. healthy individuals may have an occasional one. One such rule
is to report hypersegmentation when there are at least 5 five-
Complete Blood Count lobed neutrophils per 100WBCs or at least 1 six-lobed neutro-
Slight macrocytosis often is the earliest sign of megaloblastic phil is noted.11 Some laboratories perform a lobe count on 100
anemia. Patients with uncomplicated megaloblastic anemia neutrophils and then calculate the mean. In megaloblastic
are expected to have decreased hemoglobin and hematocrit anemia, the mean lobe count should be greater than 3.4.11 The
values, pancytopenia, and reticulocytopenia. Megaloblastic cause of the hypersegmentation is not understood, despite
anemia develops slowly, and the degree of anemia is often considerable investigation.51 More recent advances in the
severe when first detected. Hemoglobin values of less than 7 understanding of growth factors and their impact on transcrip-
or 8g/dL are not unusual.4 When the hematocrit is less than tion factors may yet solve this mystery. Nevertheless, a search
20%, erythroblasts with megaloblastic nuclei, including an for neutrophil hypersegmentation on a peripheral blood film
occasional promegaloblast, may appear in the peripheral constitutes an inexpensive yet sensitive screening test for mega-
blood. The mean cell volume (MCV) is usually 100 to 150fL loblastic anemia.
and commonly is greater than 120fL, although coexisting iron
deficiency, thalassemia trait, or inflammation can prevent mac-
rocytosis. The mean cell hemoglobin (MCH) is elevated by the Bilirubin and Lactate Dehydrogenase Levels
increased volume of the cells, but the mean cell hemoglobin Although generally considered a nutritional anemia, megalo-
concentration (MCHC) is usually within the reference range, blastic anemia is in one sense a hemolytic anemia, because the
because hemoglobin production is unaffected. The RBC distri- cells die during division in the marrow. This intramedullary
bution width (RDW) is also elevated. cell death is known as ineffective erythropoiesis because many
The characteristic morphologic findings of megaloblastic RBCs never enter the circulationhence the decrease in reticu-
anemia in the peripheral blood include oval macrocytes locytes in the peripheral blood. The usual signs of hemolysis
(enlarged oval RBCs) (Figure 20-6) and hypersegmented neu- are evident in the serum, including an elevation in the levels
trophils with six or more lobes (Figure 20-7).49 Impaired cell of total and indirect bilirubin and lactate dehydrogenase (pre-
production results in a low absolute reticulocyte count, espe- dominantly RBC derived).
cially in light of the severity of the anemia, and polychromasia The constellation of findings including macrocytic anemia,
is not seen on the peripheral blood film. Additional morpho- moderate to marked pancytopenia, reticulocytopenia, oval
logic changes may include the presence of teardrop cells, RBC macrocytes, hypersegmented neutrophils, plus increased levels
fragments, and microspherocytes. These smaller cells further of total and indirect bilirubin and lactate dehydrogenase justi-
increase the RDW. Nucleated RBCs, Howell-Jolly bodies, baso- fies further testing to confirm a diagnosis of megaloblastic
philic stippling, and Cabot rings may be observed. anemia and determine its cause. Occasionally, the classic find-
ings may be obscured by coexisting conditions such as iron
White Blood Cell Manual Differential deficiency, which makes the diagnosis more challenging. Most
Hypersegmentation of the neutrophils is essentially pathogno- hematologic aberrations do not appear until vitamin deficiency
monic for megaloblastic anemia. It appears early in the course is fairly well advanced (Box 20-4).52

Figure 20-6 Oval macrocytes, teardrop cells (dacryocytes), other red


blood cell abnormalities, and a small lymphocyte for size comparison in Figure 20-7 Hypersegmented neutrophil in megaloblastic anemia
megaloblastic anemia (peripheral blood, 500). (peripheral blood, 1000).
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 277

Figure 20-8 Erythroid precursors in megaloblastic anemia (bone marrow,


1000).

BOX 20-4 Sequence of Development of


Figure 20-9 Giant metamyelocyte in megaloblastic anemia (bone marrow,
Megaloblastic Anemias
original magnification 1000).
1. Decrease in vitamin levels
2. Hypersegmentation of neutrophils in peripheral blood
Megakaryocytes do not show consistent changes in megalo-
3. Oval macrocytes in peripheral blood
blastic anemia. They may be either increased or decreased in
4. Megaloblastosis in bone marrow
number and may show diminished lobulation. The latter
5. Anemia
finding is not consistently seen, however, and even when
present is difficult to assess.

Specific Diagnostic Tests Assays for Folate, Vitamin B12, Methylmalonic Acid,
Bone Marrow Examination and Homocysteine
Modern tests for vitamin deficiencies and autoimmune anti- Although bone marrow aspiration is confirmatory for megalo-
bodies have made bone marrow examination an infrequently blastosis, the invasiveness of the procedure and its expense
used diagnostic test for megaloblastic anemia. Nevertheless, it mean that other testing is performed more often than a bone
remains the reference confirmatory test to identify the megalo- marrow examination. Furthermore, the confirmation of mega-
blastic appearance of the developing RBCs. loblastic morphology in the marrow does not identify its cause.
Megaloblastic, in contrast to macrocytic, anemia refers to spe- Tests for serum levels of folate and vitamin B12 are readily avail-
cific morphologic changes in the developing RBCs. The cells able using immunoassay. RBC folate levels may also be mea-
are characterized by a nuclear-cytoplasmic asynchrony in which sured. Unlike serum folate levels, which fluctuate with diet,
the cytoplasm matures as expected with increasing pinkness as RBC folate values are stable and may be a more accurate reflec-
hemoglobin accumulates. The nucleus lags behind, however, tion of true folate status53; however, current RBC folate tests
appearing younger than expected for the degree of maturity of have less than optimal sensitivity and specificity and have not
the cytoplasm (Figure 20-8). This asynchrony is most striking been validated in actual patients with normal and deficient
at the stage of the polychromatophilic normoblast. The cyto- folate levels. Thus the serum folate level is preferred over RBC
plasm appears pinkish blue as expected for that stage, but the folate level in the United States as the initial test for evaluation
nuclear chromatin remains more open than expected, similar of folate deficiency.5
to that in the nucleus of a basophilic normoblast. Overall, the Some laboratories conduct a reflexive assay for methylma-
marrow is hypercellular, with a myeloid-to-erythroid ratio of lonic acid if vitamin B12 levels are low. As indicated previously,
about 1:1 by virtue of the increased erythropoietic activity. The in addition to playing a role in folate metabolism, vitamin B12
hematopoiesis is ineffective, however, and although cell pro- is a cofactor in the conversion of methylmalonyl CoA to suc-
duction in the bone marrow is increased, the death of cells in cinyl CoA by the enzyme methylmalonyl CoA mutase (see
the marrow results in peripheral pancytopenia. Figure 20-2). If vitamin B12 is deficient, methylmalonyl CoA
The WBCs are also affected in megaloblastic anemia and accumulates. Some of it hydrolyzes to methylmalonic acid, and
appear larger than normal. This is most evident in metamyelo- the increase can be detected in serum and urine. Because meth-
cytes and bands, because in the usual development of neutro- ylmalonic acid is also elevated in patients with impaired renal
phils, the cells should be getting smaller at these stages. The function, increased levels cannot be definitively related to
effect creates what are called giant metamyelocytes (Figure vitamin B12 deficiency.25 Methylmalonic acid is assayed by gas
20-9) and bands. chromatographytandem mass spectrometry.
278 PART IV Hematopathology: Erythrocyte Disorders

Homocysteine levels are affected by folate and vitamin B12. Holotranscobalamin Assay
5-Methyl THF donates a methyl group to homocysteine in Holotranscobalamin is the metabolically active form of vitamin
the generation of methionine. This process uses vitamin B12 B12. Until recently, methods for measuring holotranscobalamin
as a coenzyme (see Figure 20-4). Thus, a deficiency in either were manual and not suitable for use in clinical laboratories.
folate or vitamin B12 results in elevated levels of homocysteine. Newer, more rapid immunoassays using monoclonal anti
Total homocysteine can be measured in either plasma or bodies specific for holotranscobalamin have been developed in
serum. Homocysteine may be assayed by gas chromatography the past several years that are both sensitive and specific.54,55
mass spectrometry, high-performance liquid chromatography,
or fluorescence polarization immunoassay. Homocysteine Deoxyuridine Suppression Test
levels are also elevated in patients with renal failure and The principle of the deoxyuridine suppression test is that the
dehydration. preincubation of normal bone marrow with deoxyuridine will
suppress the subsequent incorporation of labeled thymidine
Gastric Analysis into DNA, because the normal cells can successfully methylate
Gastric analysis may be used to confirm achlorhydria, an the uridine into thymidine. However, in patients with either a
expected finding in pernicious anemia. Achlorhydria occurs in vitamin B12 or a folate deficiency, this suppression is abnormally
other conditions, however, including natural aging. When low. By adding either vitamin B12 or folate to the test cells, one
other causes of vitamin B12 deficiency have been eliminated, a can determine whether the inadequate suppression is caused by
finding of achlorhydria is supportive, although not diagnostic, vitamin B12 or folate deficiency.56-58 Although micromethods have
of pernicious anemia. The H+ concentration is determined by been developed, the necessity of using bone marrow tissue and
pH measurement. the complexity of the test make it impractical for clinical testing.

Antibody Assays Stool Analysis for Parasites


Antibodies to intrinsic factor and parietal cells can be detected When vitamin B12 is found to be deficient, a stool analysis for
in the serum of most patients with pernicious anemia. Various eggs or proglottids of the fish tapeworm D. latum may be part
immunoassays can detect intrinsic factorblocking antibodies; of the diagnostic workup.
parietal cell antibodies can be detected by indirect fluorescent See Table 20-1 for a summary of laboratory tests used to
antibody techniques or enzyme-linked immunosorbent assays. diagnose vitamin B12 and folate deficiency.

TABLE 20-1 Laboratory Tests Used to Diagnose Vitamin B12 and Folate Deficiency
Folate Deficiency Vitamin B12 Deficiency
Screening tests Complete blood count Hb, Hct, RBCs, WBCs, PLTs Same as Folate Deficiency
MCV, MCH
Manual differential count Hypersegmented neutrophils, oval macrocytes, Same as Folate Deficiency
anisocytosis, poikilocytosis, RBC inclusions
Absolute reticulocyte count
Serum total and indirect bilirubin
Serum lactate dehydrogenase
Specific diagnostic Bone marrow examination* Erythroid hyperplasia (ineffective) Same as Folate Deficiency
tests Presence of megaloblasts
Serum vitamin B12 N
Serum folate N or
RBC folate N or
Serum methylmalonic acid N
Serum/plasma homocysteine
Antibodies to intrinsic factor and Absent Present in pernicious anemia
gastric parietal cells
Gastric analysis* N Achlorhydria in pernicious anemia
Holotranscobalamin assay N
Stool analysis for parasites Negative Diphyllobothrium latum may be
the cause of deficiency

, Increased; , decreased; Hb, hemoglobin; Hct, hematocrit; MCH, mean cell hemoglobin; MCV, mean cell volume; N, within reference range; PLT, platelet; RBC, red blood
cell; WBC, white blood cell.
*Bone marrow examination and gastric analysis are not usually required for diagnosis.

Without vitamin B12, the cell is unable to produce intracellular polyglutamated tetrahydrofolate; therefore, 5-methyltetrahydrofolate leaks out of the cell, which results in a decreased
level of intracellular folate.

Holotranscobalamin level is also decreased in transcobalamin deficiency.


CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 279

MCV  100 fL

Normal newborns
Reticulocytosis Megaloblastic characteristics
Liver disease No Oval macrocytes
Hypersegmented neutrophils
Chronic alcoholism Pancytopenia
Bone marrow failure

Yes

Serum Folate and Vitamin B12 Levels

Vitamin B12 Deficiency Folate Deficiency No Deficiency

Inadequate intake Inadequate intake Myelodysplastic syndrome


Increased need Increased need Acute erythroleukemia
Impaired absorption impaired absorption Congenital dyserythropoietic anemia
Inability to split B12 from food Impaired use Reverse transcriptase inhibitors
Inability to split B12 from haptocorrin
Lack of intrinsic factor Excessive loss (renal dialysis)
Pernicious anemia
H. pylori infection
Gastrectomy
Hereditary intrinsic factor deficiency
General malabsorption
Inherited errors in transport/absorption
Competition for vitamin B12
D. latum infection
Blind loop syndrome

Figure 20-10 Algorithm for preliminary investigation of macrocytic anemias. MCV, Mean cell volume.

intramuscularly to treat pernicious anemia to bypass the need


MACROCYTIC NONMEGALOBLASTIC
for intrinsic factor. Large doses of oral vitamin B12 have been
ANEMIAS
proposed to be effective in the treatment of pernicious
The macrocytic nonmegaloblastic anemias are macrocytic anemia40,59; however, this treatment has not been validated in
anemias in which DNA synthesis is unimpaired. The macrocy- clinical practice. Folic acid can be administered orally. The
tosis tends to be mild; the MCV usually ranges from 100 to inappropriate treatment of vitamin B12 deficiency with folic
110fL and rarely exceeds 120fL. Patients with nonmegalo- acid improves the anemia, but does not correct or stop the
blastic, macrocytic anemia lack hypersegmented neutrophils progress of the neurologic damage, which may advance to an
and oval macrocytes in the peripheral blood and megaloblasts irreversible state.25 Thus proper diagnosis prior to treatment
in the bone marrow. Macrocytosis may be physiologically is important. Iron is often supplemented concurrently to
normal, as in the newborn (see Chapter 38), or the result of support the rapid cell production that accompanies effective
pathology, as in liver disease, chronic alcoholism, or bone treatment.
marrow failure. Reticulocytosis is a common cause of macro- When proper treatment is initiated, the bodys response is
cytosis. Figure 20-10 presents an algorithm for the preliminary prompt and brisk and can be used to confirm the accuracy of
investigation of macrocytic anemias. the diagnosis. The bone marrow morphology will begin to
revert to a normoblastic appearance within a few hours of
treatment. A substantial reticulocyte response is apparent at
TREATMENT
about 1 week, with hemoglobin increasing toward normal
Treatment should be directed at the specific vitamin deficiency levels in about 3 weeks.11 Hypersegmented neutrophils disap-
established by the diagnostic tests and should include address- pear from the peripheral blood within 2 weeks.5 Thus, with
ing the cause of the deficiency (e.g., better nutrition, treatment proper treatment, hematologic parameters may return to
for D. latum), if possible. Vitamin B12 is administered normal within 3 to 6 weeks.
280 PART IV Hematopathology: Erythrocyte Disorders

SUMMARY
Impaired DNA synthesis affects all rapidly dividing cells of the bacteria in intestinal blind loops. Lack of intrinsic factor may result
body, including the skin, gastrointestinal tract, and bone marrow. from loss of gastric parietal cells with pernicious anemia, H. pylori
The effect on hematologic cells results in megaloblastic anemia, infection, gastrectomy, or inherited intrinsic factor deficiency.
so named for the very large RBCs that develop in the bone marrow. Pernicious anemia is vitamin B12 deficiency resulting from an
Vitamin B12 and folate are needed for the production of thymidine autoimmune disease that causes atrophic destruction of gastric
nucleotides for DNA synthesis. Deficiencies of either vitamin parietal cells. H+ and intrinsic factor secretion is lost. Antibodies
impair DNA replication, halt cell division, and increase apoptosis, to parietal cells or intrinsic factor or both are detectable in the
which results in ineffective erythropoiesis and megaloblastic serum.
morphology. Classic CBC findings in megaloblastic anemia include decreased
Patients with vitamin B12 or folate deficiency develop megaloblas- hemoglobin level, hematocrit, and RBC count; leukopenia; throm-
tic anemia and the general symptoms of anemia. Patients with bocytopenia; decreased absolute reticulocyte count; elevated MCV
vitamin B12 deficiency may develop neuropathies and neuropsy- (usually greater than 120fL); elevated RDW and MCH; MCHC
chiatric abnormalities as a result of demyelinization of nerves within the reference range; oval macrocytes; and hypersegmented
in the peripheral and central nervous system. Folate deficiency, neutrophils. Additional abnormal laboratory test findings may
in particular, leads to elevation of homocysteine levels and pos- include elevated levels of total and indirect serum bilirubin and
sible risk of coronary artery disease. Peripheral neuropathy and lactate dehydrogenase due to the ineffective erythropoiesis and
depression also may accompany folate deficiency. Folate defi- intramedullary hemolysis.
ciency in early pregnancy can lead to neural tube defects in The bone marrow in megaloblastic anemia is hyperplastic with
the fetus. increased erythropoiesis; however, it is ineffective due to increased
Folate deficiency may result from inadequate intake, increased apoptosis of developing cells. RBC precursors show nuclear-
need with growth or pregnancy, impaired absorption, impaired cytoplasmic asynchrony, with the nuclear maturation lagging
use, or excessive loss. The action of folates can be impaired by behind the cytoplasmic maturation. Giant metamyelocytes and
drugs such as those used to treat epilepsy or cancer. Renal dialy- bands are evident.
sis patients experience significant folate loss to the dialysate. The cause of megaloblastic anemia is determined using specific
Vitamin B12 deficiency arises from inadequate intake, increased immunoassays for serum and RBC folate and serum vitamin B12.
need, or inadequate absorption. Inadequate intake of vitamin B12, Immunoassays for antibodies to intrinsic factor and parietal cells
although possible, is uncommon, because vitamin B12 is ubiqui- can aid in the diagnosis of pernicious anemia. Additional tests for
tous in animal products. Pregnancy, lactation, and growth create gastrointestinal disease or parasites may be needed.
increased need for vitamin B12. Treatment of megaloblastic anemia is directed at correcting
Absorption of vitamin B12 depends on production of intrinsic factor the cause of the deficiency, supplementing the missing vitamin,
by parietal cells of the stomach. or both.
Impaired absorption of vitamin B12 can be caused by several For pernicious anemia, lifelong supplementation with vitamin B12
mechanisms. Decrease in gastric acid production or lack of trypsin is necessary.
in the intestine causes vitamin B12 to be excreted in the stool rather
than absorbed. Malabsorption can be caused by intestinal dis- Now that you have completed this chapter, go back and
eases, such as inflammatory bowel disease. Competition for read again the case study at the beginning and respond
vitamin B12 can develop from an intestinal parasite (D. latum) or to the questions presented.

R E V I E W Q UESTIONS
1. Which of the following findings is consistent with a diag- 3. Which one of the following statements characterizes the
nosis of megaloblastic anemia? relationships among macrocytic anemia, megaloblastic
a. Hyposegmentation of neutrophils anemia, and pernicious anemia?
b. Decreased serum lactate dehydrogenase level a. Macrocytic anemias are megaloblastic.
c. Absolute increase in reticulocytes b. Macrocytic anemia is pernicious anemia.
d. Increased MCV c. Megaloblastic anemia is macrocytic.
d. Megaloblastic anemia is pernicious anemia.
2. A patient has a clinical picture of megaloblastic anemia.
The serum folate level is decreased, and the serum vitamin 4. Which of the following CBC findings is most suggestive of
B12 level is normal. What is the expected value for the a megaloblastic anemia?
methylmalonic acid assay? a. MCV of 103fL
a. Increased b. Hypersegmentation of neutrophils
b. Decreased c. RDW of 16%
c. Normal d. Hemoglobin concentration of 9.1g/dL
CHAPTER 20 Anemias Caused by Defects of DNA Metabolism 281

5. In the following description of a bone marrow smear, find 8. Folate and vitamin B12 work together in the production of:
the statement that is inconsistent with the expected picture a. Amino acids
in megaloblastic anemia. b. RNA
The marrow appears hypercellular with a myeloid-to- c. Adenosine triphosphate
erythroid ratio of 1:1 due to prominent erythroid hyper- d. DNA
plasia. Megakaryocytes appear normal in number and
appearance. The WBC elements appear larger than normal 9. The macrocytosis associated with megaloblastic anemia
with especially large metamyelocytes, although they results from:
otherwise appear morphologically normal. The RBC pre- a. Reduced numbers of cell divisions
cursors also appear large. There is nuclear-cytoplasmic b. Activation of a gene that is typically active only in
asynchrony, with the nucleus appearing more mature than megakaryocytes
expected for the color of the cytoplasm. c. Reduced concentration of hemoglobin in the cells so
a. Erythroid nuclei that are more mature than cytoplasm that larger cells are needed to provide the same
b. Larger than normal WBC elements oxygen-carrying capacity
c. Larger than normal RBCs d. Increased production of reticulocytes in an attempt to
d. Normal appearance of megakaryocytes compensate for the anemia

6. Which of the following findings would be inconsistent with 10. Individuals of which one of the following groups are at
elevated titers of parietal cell antibodies? highest risk for pernicious anemia?
a. Hypersegmentation of neutrophils a. Malnourished infants
b. Low levels of methylmalonic acid b. Children during growth periods
c. Macrocytic RBCs c. Persons older than 60 years of age
d. Elevated levels of intrinsic factorblocking antibodies d. Pregnant women

7. Which of the following is the most metabolically active


form of absorbed vitamin B12?
a. Transcobalamin
b. Intrinsic factorvitamin B12 complex
c. Holotranscobalamin
d. Haptocorrinvitamin B12 complex

REFERENCES
1. Scott JM: Folate and vitamin B12. Proc Nutr Soc 58:441-448, 10. Geen D, Sudre P, Anwar D, et al: Causes of macrocytosis in
1999. HIV-infected patients not treated with zidovudine. Swiss HIV
2. Wickramasinghe SN: The wide spectrum and unresolved Cohort Study. J Infect 40:160-163, 2000.
issues of megaloblastic anemia. Semin Hematol 36:3-18, 1999. 11. Carmel R, Rosenblatt DS: Disorders of cobalamin and folate
3. McPartlin J: Folic acid. In Caballero B, Allen L, Prentice A, metabolism. In Handin RI, Lux SE, Stossel TP, editors: Blood:
editors: Encyclopedia of human nutrition, ed 2, Boston, 2005, principles and practice of hematology, ed 2, Philadelphia, 2003,
Elsevier/Academic Press, pp 257-264. Lippincott Williams & Wilkins, pp 1361-1398.
4. Carmel R: Megaloblastic Anemias: Disorders of Impaired 12. Solomon LR: Disorders of cobalamin (vitamin B12) metabo-
DNA Synthesis. In Greer JP, Foerster J, Rodgers GM, et al, lism: emerging concepts in pathophysiology, diagnosis and
editors: Wintrobes clinical hematology, ed 12, Philadelphia, treatment. Blood Rev 21:113-130, 2007.
2009, Wolters Kluwer Health/Lippincott Williams & Wilkins, 13. Scalabrino G, Peracchi M: New insights into the patho
pp 1143-1172. physiology of cobalamin deficiency. Trends Mol Med
5. Antony AC: Megaloblastic anemias. In Hoffman R, Benz 12:247-254, 2006.
EJ, Shattil SJ, et al, editors: Hematology: basic principles and 14. Morrison HI, Schaubel D, Desmeules M, et al: Serum folate
practice, ed 5, Philadelphia, 2009, Churchill Livingstone, and risk of fatal coronary heart disease. JAMA 275:1893-
pp 491-524. 1896, 1996.
6. Goulian M, Bleile B, Tseng BY: Methotrexate-induced misin- 15. Voutilainen S, Virtanen JK, Rissanen TH, et al: Serum folate
corporation of uracil into DNA. Proc Natl Acad Sci U S A and homocysteine and the incidence of acute coronary
77:1956-1960, 1980. events: the Kuopio Ischaemic Heart Disease Risk Factor
7. Wickramasinghe SN, Fida S: Bone marrow cells from vitamin Study. Am J Clin Nutr 80:317-323, 2004.
B12- and folate-deficient patients misincorporate uracil into 16. de Bree A, Verschuren WM, Blom HJ, et al: Coronary heart
DNA. Blood 83:1656-1661, 1994. disease mortality, plasma homocysteine, and B-vitamins: a
8. Koury MJ, Horne DW, Brown ZA, et al: Apoptosis of late stage prospective study. Atherosclerosis 166:369-377, 2003.
erythroblasts in megaloblastic anemia: association with DNA 17. Hung J, Beilby JP, Knuiman MW, et al: Folate and vitamin
damage and macrocyte production. Blood 89:4617-4623, B12 and risk of fatal cardiovascular disease: cohort study from
1997. Busselton, Western Australia. BMJ 326:131-137, 2003.
9. Das KC, Das M, Jadaon MM, et al: Megaloblastosis: from 18. Soinio M, Marniemi J, Laakso M, et al: Elevated plasma
morphos to molecules. Med Princ Pract 14:2-14, 2006. homocysteine level is an independent predictor of coronary
282 PART IV Hematopathology: Erythrocyte Disorders

heart disease events in patients with type 2 diabetes mellitus. 39. Seetharam B, Bose S, Li N: Cellular import of cobalamin
Ann Intern Med 140:94-100, 2004. (vitamin B-12). J Nutr 129:1761-1764, 1999.
19. Lindeman RD, Romero LJ, Yau CL, et al: Serum homocysteine 40. Dali-Youcef N, Andres E: An update on cobalamin deficiency
concentrations and their relation to serum folate and vitamin in adults. QJM 102:17-28, 2009.
B12 concentrations and coronary artery disease prevalence in 41. Toh BH, van Driel IR, Gleeson PA: Pernicious anemia. N Engl
an urban, bi-ethnic community. Ethn Dis 13:178-185, 2003. J Med 337:1441-1448, 1997.
20. Bottiglieri T, Laundry M, Crellin R, et al: Homocysteine, 42. Karlsson FA, Burman P, Loof L, et al: Major parietal cell
folate, methylation, and monoamine metabolism in depres- antigen in autoimmune gastritis with pernicious anemia in
sion. J Neurol Neurosurg Psychiatry 69:228-232, 2000. the acid-producing H+,K+-adenosine triphosphatase of the
21. Kronenberg G, Colla M, Endres M: Folic acid, neurodegenera- stomach. J Clin Invest 81:475-479, 1988.
tive and neuropsychiatric disease. Curr Mol Med 9:315-323, 43. Bardhan KD, Hall JR, Spray GH, et al: Blocking and binding
2009. autoantibody to intrinsic factor. Lancet 1:62-64, 1968.
22. Reynolds EH, Rothfeld P, Pincus JH: Neurological disease 44. Strickland RG, Hooper B: The parietal cell heteroantibody in
associated with folate deficiency. BMJ 2:398-400, 1973. human sera: prevalence in a normal population and relation-
23. Alpert JE, Mischoulon D, Nierenberg AA, et al: Nutrition and ship to parietal cell autoantibody. Pathology 4:259-263, 1972.
depression: focus on folate. Nutrition 16:544-546, 2000. 45. Kaferle J, Strzoda CE: Evaluation of macrocytosis. Am Fam
24. Blom HJ, Shaw GM, den Heijer M, et al: Neural tube defects Physician 79(3):203-208, 2009.
and folate: case far from closed. Nat Rev Neurosci 7:724-731, 46. Kaptan K, Beyan C, Ural AU, et al: Helicobacter pyloriis it a
2006. novel causative agent in vitamin B12 deficiency? Arch Intern
25. Scott JM, Browne P: Megaloblastic anemia. In Caballero B, Med 160:1349-1353, 2000.
Allen L, Prentice A, editors: Encyclopedia of human nutrition, 47. Morel CF, Watkins D, Scott P, et al: Prenatal diagnosis for
ed 2, Boston, 2005, Elsevier/Academic Press, pp 109-117. methylmalonic acidemia and inborn errors of vitamin B12
26. Gallagher ML: Vitamins. In Mahan LK, Escott-Stump S, metabolism and transport. Mol Genet Metab 86:160-171,
editors: Krauses food, nutrition, and diet therapy, ed 11, 2005.
Philadelphia, 2004, Saunders, pp 74-119. 48. Nyberg W, Grsbeck R, Saarni M, et al: Serum vitamin B12
27. Qui A, Jansen M, Sakaris A, et al: Identification of an intesti- levels and incidence of tape worm anemia in a population
nal folate transporter and the molecular basis for hereditary heavily infected with Diphyllobothrium latum. Am J Clin Nutr
folate malabsorption. Cell 127:917-928, 2006. 9:606-612, 1961.
28. Dixon KH, Lanpher BC, Chiu J, et al: A novel cDNA restores 49. Zittoun J, Zittoun R: Modern clinical testing strategies in
reduced folate carrier activity and methotrexate sensitivity to cobalamin and folate deficiency. Semin Hematol 36:35-46,
transport deficient cells. J Biol Chem 269(1):17-20, 1994. 1999.
29. Nath SK: Tropical Sprue. Curr Gastroenterol Rep 7:343-349, 50. Herbert V: Experimental nutritional folate deficiency in man.
2005. Trans Assoc Am Physicians 75:307-320, 1962.
30. Waxman S, Metz J, Herbert V: Defective DNA synthesis in 51. Wickramasinghe SN: The wide spectrum and unresolved
human megaloblastic bone marrow: effects of homocysteine issues in megaloblastic anemia. Semin Hematol 36:3-18,
and methionine. J Clin Invest 48:284-289, 1969. 1999.
31. Moran RG, Keyomarsi K: Biochemical rationale for the 52. Carmel R: Introduction: beyond megaloblastic anemia. Semin
synergism of 5-fluorouracil and folinic acid. NCI Monogr Hematol 36:1-2, 1999.
5:159-163, 1987. 53. Piyathilake CJ, Robinson CB, Cornwell P: A practical approach
32. Kishi T, Fujita N, Eguchi T, et al: Mechanism for reduction of to red blood cell folate analysis. Anal Chem Insights 2:107-
serum folate by antiepileptic drugs during prolonged therapy. 110, 2007.
J Neurol Sci 145:109-112, 1997. 54. Brady J, Wilson L, McGregor L, et al: Active B12: a rapid, auto-
33. Froscher W, Maier V, Laage M, et al: Folate deficiency, anti- mated assay for holotranscobalamin on the Abbott AxSYM
convulsant drugs, and psychiatric morbidity. Clin Neurophar- analyzer. Clin Chem 54(3):567-573, 2008.
macol 18:165-182, 1995. 55. Ulleland M, Eilertsen I, Quadros EV, et al: Direct assay for
34. Green R: Cobalamins. In Caballero B, Allen L, Prentice A, cobalamin bound to transcobalamin (holo-transcobalamin)
editors: Encyclopedia of human nutrition, ed 2, Boston, 2005, in serum. Clin Chem 48(3):526-532, 2002.
Elsevier/Academic Press, pp 401-407. 56. Metz J: The deoxyuridine suppression test. CRC Crit Rev Clin
35. He Q, Madsen M, Kilkenney A, et al: Amnionless function is Lab Sci 20:205-241, 1984.
required for cubilin brush-border expression and intrinsic 57. Wickramasinghe SN, Matthew JH: Deoxyuridine suppress:
factor-cobalamin (vitamin B12) absorption in vivo. Blood biochemical basis and diagnostic applications. Blood Rev
106:1447-1453, 2005. 2:168-177, 1988.
36. Morel CF, Rosenblatt DS. In Sarafoglou K, Hoffman GF, Roth 58. Carmel R, Bedros AA, Mace JW, et al: Congenital methylma-
KS, editors: Pediatric endocrinology and inborn errors of metabo- lonic aciduriahomocystinuria with megaloblastic anemia:
lism, New York, 2009, McGraw-Hill, pp 195-210. observation on response to hydroxycobalamin and on the
37. Fyfe JC, Madsen M, Hojrup P, et al: The functional cobalamin effect of homocysteine and methionine on the deoxyuridine
(vitamin B12)intrinsic factor receptor is a novel complex of suppression test. Blood 55:570-579, 1980.
cubilin and amnionless. Blood 103:1573-1579, 2004. 59. Butler CC, Vidal-Alaball J, Cannings-John R, et al: Oral
38. Afman LA, Van Der Put NMJ, Thomas CMG, et al: Reduced vitamin B12 versus intramuscular vitamin B12 for vitamin B12
vitamin B12 binding by transcobalamin II increases the risk deficiency: a systematic review of randomized controlled
of neural tube defects. QJM 94:159-166, 2001. trials. Fam Pract 23:279-285, 2006.
Bone Marrow Failure
Elaine M. Keohane
21
OUTLINE OBJECTIVES
Pathophysiology of Bone After completion of this chapter, the reader will be able to:
Marrow Failure
Aplastic Anemia 1. Describe the clinical consequences of bone marrow 7. Differentiate among causes of pancytopenia based
Acquired Aplastic Anemia failure. on laboratory tests and clinical findings.
Inherited Aplastic Anemia 2. Describe the etiology of acquired and inherited 8. Discuss the possible relationship between defects in
Differential Diagnosis aplastic anemias. the telomerase complex and bone marrow failure in
Other Forms of Bone 3. Discuss the pathophysiologic mechanisms of acquired and inherited aplastic anemia.
Marrow Failure acquired and inherited aplastic anemias. 9. Compare and contrast the pathophysiology, clinical
Pure Red Cell Aplasia 4. Describe the characteristic peripheral blood and picture, and laboratory findings in transient erythro-
Congenital bone marrow features in aplastic anemia. blastopenia of childhood, Diamond-Blackfan anemia,
Dyserythropoietic 5. Classify aplastic anemia as nonsevere, severe, or and congenital dyserythropoietic anemia.
Anemia
very severe based on laboratory tests. 10. Describe the mechanisms causing cytopenia in
Myelophthisic Anemia
6. Discuss treatment modalities for acquired and myelophthisic anemia and anemia of chronic kidney
Anemia of Chronic Kidney
Disease inherited aplastic anemia and the patients for whom disease.
each is most appropriate.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 52-year-old female data entry clerk complained of bilateral Serum vitamin B12 and folate levels were normal. Bone
wrist pain. Her physician prescribed a nonsteroidal anti marrow aspirate was normocellular with dyserythropoiesis,
inflammatory agent. Her wrist pain improved; however, over but normal myelopoiesis and megakaryopoiesis. Iron stain
the next 3 months, she noted increasing fatigue and scattered revealed normal stores. Bone marrow biopsy specimen was
bruises. Past medical history was unremarkable. She was moderately hypocellular (30%) with a reduction in all three
taking no other medications and had no recent chemical cell lines. There was no increase in reticulin or blasts. Cyto-
exposure. Physical examination revealed pallor and scattered genetic testing revealed a normal karyotype, and results of
ecchymoses with petechiae on her chest and shoulders with flow cytometry for PNH cells was negative.
no other abnormalities. Complete blood count results were 1. What term is used to describe a decrease in all cell lines
as follows: in the peripheral blood?
2. Which anemia of bone marrow failure should be
Patient Reference Range
considered?
9
WBCs ( 10 /L) 2.0 4.5-11.0 3. How would an increase in either reticulin or blasts alter
Hb (g/dL) 8.0 12.0-15.0 the preliminary diagnosis?
MCV (fL) 104 80-100 4. How would the severity of this patients condition be
Platelets ( 109/L) 27 150-450 classified?
Reticulocytes (%) 0.6% 0.5-1.5 5. What treatment modality would be considered for this
Reticulocytes ( 109/L) 16 25-75 patient?
Neutrophils ( 109/L) 1.1 2.3-8.1
Lymphocytes ( 109/L) 0.4 0.8-4.8

283
284 PART IV Hematopathology: Erythrocyte Disorders

PATHOPHYSIOLOGY OF BONE BOX 21-1 Characteristic Features of Aplastic Anemia


MARROW FAILURE Peripheral blood pancytopenia
Bone marrow failure is the reduction or cessation of blood cell Reticulocytopenia
production affecting one or more cell lines. Pancytopenia, or Bone marrow hypocellularity
decreased numbers of circulating red blood cells (RBCs), white Depletion of hematopoietic stem cells
blood cells (WBCs), and platelets, is seen in most cases of bone
marrow failure, particularly in severe or advanced stages.
The pathophysiology of bone marrow failure includes the
following mechanisms: (1) destruction of hematopoietic stem BOX 21-2 Etiologic Classification of Aplastic Anemia
cells due to injury by drugs, chemicals, radiation, viruses, or
autoimmune mechanisms; (2) premature senescence and apop- Acquired (80-85% of cases)
tosis of hematopoietic stem cells due to mutations; (3) ineffec- Idiopathic (70% of acquired cases)
tive hematopoiesis due to stem cell mutations or vitamin B12 Secondary (10-15% of acquired cases)
or folate deficiency; (4) disruption of the bone marrow micro- Dose dependent/predictable
environment that supports hematopoiesis; (5) decreased pro- Cytotoxic drugs
duction of hematopoietic growth factors or related hormones; Benzene
and (6) loss of normal hematopoietic tissue due to infiltration Radiation
of the marrow space with abnormal cells. Idiosyncratic
The clinical consequences of bone marrow failure vary Drugs (see Box 21-3)
depending on the extent and duration of the cytopenias. Severe Chemicals
pancytopenia can be rapidly fatal if untreated. Some patients Insecticides
may present initially with no symptoms, and their cytopenia Cutting/lubricating oils
may be inadvertently detected during a routine examination. Viruses
Thrombocytopenia can result in clinically significant bleeding. Epstein-Barr virus
The decrease in RBCs and hemoglobin (Hb) leads to symptoms Hepatitis virus (non-A, non-B, non-C, non-G)
of anemia, including fatigue, pallor, and cardiovascular comp Human immunodeficiency virus
lications. Sustained neutropenia increases the risk of bacterial Miscellaneous conditions
or fungal infections that can be life-threatening. Paroxysmal nocturnal hemoglobinuria
This chapter focuses on aplastic anemia, a bone marrow Autoimmune diseases
failure syndrome resulting from damaged or defective stem Pregnancy
cells (mechanisms 1 and 2 listed earlier). Bone marrow failure
resulting from other mechanisms may have a presentation Inherited (15-20% of cases)
similar to that of aplastic anemia and differentiation is dis- Fanconi anemia
cussed later. Because there are many mechanisms involved Dyskeratosis congenita
in the various bone marrow failure syndromes, accurate diag- Shwachman-Diamond syndrome
nosis is essential so that the appropriate treatment can be
instituted.
to 15% are secondary.3 Idiopathic and secondary aplastic
anemia have similar clinical and laboratory findings. Initially,
APLASTIC ANEMIA
patients may manifest a macrocytic or normocytic anemia with
Aplastic anemia is a rare but potentially fatal bone marrow reticulocytopenia. Depending on the progression of the bone
failure syndrome. In 1888, Ehrlich provided the first case marrow failure, pancytopenia may develop slowly or progress
report of aplastic anemia involving a patient with severe at a rapid rate with complete cessation of hematopoiesis.
anemia, neutropenia, and a yellow hypocellular marrow on
postmortem examination.1 The name aplastic anemia was given Incidence
to the disease by Vaquez and Aubertin in 1904.2 The charac- In North America and Europe, the incidence per year approxi-
teristic features of aplastic anemia include pancytopenia, retic- mates 2 in 1 million.6 In Asia and East Asia, the incidence is
ulocytopenia, bone marrow hypocellularity, and depletion of two to three times higher than in North America or Europe,
hematopoietic stem cells (Box 21-1). Approximately 80% to which may be due to environmental or genetic differences.7
85% of cases of aplastic anemias are acquired, and 15% to 20% Aplastic anemia can occur at any age, with the highest fre-
are inherited.3 Box 21-2 provides an etiologic classification.3-5 quency at 15 to 25 years and the second highest frequency over
the age of 60.4,6,8 There is no gender difference in incidence.6
Acquired Aplastic Anemia
Acquired aplastic anemia is classified as idiopathic when the Etiology
cause is unknown and as secondary when the cause can be The cause of the bone marrow failure in idiopathic aplastic
identified. Approximately 70% of cases are idiopathic and 10% anemia is unknown. Secondary aplastic anemia is associated
CHAPTER 21 Bone Marrow Failure 285

with exposure to certain drugs, chemicals, radiation, or infec-


tious agents. Cytotoxic drugs, radiation, and benzene suppress BOX 21-3 Selected Drugs Reported to Have a Rare
the bone marrow in a predictable, dose-dependent manner.4,5 Association with Idiosyncratic Aplastic Anemia
Depending on the dose and exposure time, the bone marrow
generally recovers after withdrawal of the agent. About 90% of Antiarthritics
cases of secondary aplastic anemia occur due to idiosyncratic Gold compounds
reactions to drugs or chemicals. In idiosyncratic reactions, the Penicillamine
bone marrow failure is unpredictable and unrelated to dose,
and the bone marrow does not usually recover when the agent Antibiotics
is withdrawn.4 Documentation of an identifiable factor or Chloramphenicol
agent inducing aplastic anemia in these cases is difficult, Sulfonamides
because evidence is primarily circumstantial and symptoms
may occur months or years after exposure. Some drugs associ- Anticonvulsants
ated with idiosyncratic, acquired aplastic anemia are listed in Carbamazepine
Box 21-3.4,8 Hydantoins
Aplastic anemia as an idiosyncratic adverse reaction to a Phenacemide
drug, chemical, or other agent is a rare event and is likely due
to a combination of genetic and environmental factors in Antidepressants
susceptible individuals. Currently no tests are available to pre Dothiepin
dict individual susceptibility to these idiosyncratic reactions; Phenothiazine
however, genetic variations in immune response pathways or
metabolic enzymes may play a role.4 There is an approximately Antidiabetic Agents
twofold higher incidence of HLA-DR2 and its major serologic Chlorpropamide
split, HLA-DR15, in aplastic anemia patients than in the Tolbutamide
general population, but the relationship of this finding to the Carbutamide
disease pathophysiology has not been elucidated.9,10 Genetic
polymorphisms in enzymes that metabolize benzene may Antiinflammatories (nonsteroidal)
increase an individuals susceptibility to its toxicity, even at low Aspirin
levels of exposure.4,11 These include polymorphisms in gluta- Diclofenac
thione S-transferase (GST) enzymes (GSTT1 and GSTM1), Fenbufen
myeloperoxidase, nicotinamide adenine dinucleotide phos- Fenoprofen
phate (reduced form):quinine oxidoreductase 1, and cyto- Ibuprofen
chrome oxidase P450 2E1.11 A deficiency in GST due to the Indomethacin
GSTT1 null genotype is overrepresented in white, Hispanics, Naproxen
and Asians with aplastic anemia with a frequency of 30%, 28%, Phenylbutazone
and 75%, respectively.12 White patients with aplastic anemia Piroxicam
also have a higher frequency (22%) of the GSTM1/GSTT1 null Sulindac
genotype than the general population.12 GST is important for
metabolism of chemical toxins, and deficiencies of this enzyme Antiprotozoals
may increase the risk of developing aplastic anemia. Further Chloroquine
study is required to assess how these genetic variations, and Quinacrine
other factors yet to be discovered, contribute to the bone
marrow suppression in aplastic anemia. Antithyroidals
Acquired aplastic anemia occurs occasionally as a complica- Methimazole
tion of infection with Epstein-Barr virus, human immunodefi- Methylthiouracil
ciency virus (HIV), hepatitis virus, and human parvovirus B19.4
A history of acute non-A, non-B, or non-C hepatitis 1 to 3 Carbonic Anhydrase Inhibitors
months before onset of pancytopenia is found in 2% to 10% Methazolamide
of patients with acquired aplastic anemia.13 Posthepatitis aplas- Mesalazine
tic anemia syndrome has a poor prognosis. Acetazolamide
Aplastic anemia associated with pregnancy is a rare occur-
rence, with fewer than 100 cases reported in the literature.14
Approximately 10% of individuals with acquired aplastic Pathophysiology
anemia have a concomitant autoimmune disease,15 and approx- The primary lesion in acquired aplastic anemia is a quantitative
imately 10% develop hemolytic or thrombotic manifestations and qualitative deficiency of hematopoietic stem cells. In
of paroxysmal nocturnal hemoglobinuria (PNH).16 The overlap culture, stem cells of patients with acquired aplastic anemia
between acquired aplastic anemia and PNH is discussed later. have diminished colony formation.17 The hematopoietic stem
286 PART IV Hematopathology: Erythrocyte Disorders

and early progenitor cell compartment is identified by expres- an immune response that cross-reacts with self-antigens.24,28
sion of CD34 surface antigens. When measured by flow cyto With regard to the latter mechanism, CD4+CD25+FOXP3+ regu-
metry, the CD34+ cell population in the bone marrow of latory T cells are decreased in aplastic anemia.33 Regulatory T
patients with aplastic anemia can be 10 times lower than that cells normally suppress autoreactive T cells, and a deficit of
in healthy individuals.17 In addition, these CD34+ cells have an these cells may facilitate an autoimmune reaction. In addition,
increased expression of Fas receptors that mediate apopto- 36.9%, 10.9%, 21.9%, and 43.8% of individuals with aplastic
sis18,19 and an increased expression of apoptosis-related genes anemia have the single nucleotide polymorphisms IFN-/+874
as determined by microarray analysis.20 The bone marrow TT, TNF-/308 AA, transforming growth factor-1/509 TT,
stromal cells are functionally normal in acquired aplastic and interleukin-6/174 GG, respectively.34 These polymor-
anemia. They produce normal or increased quantities of growth phisms result in cytokine overproduction and may impart a
factors, and they are able to support the growth of CD34+ cells genetic susceptibility to aplastic anemia and contribute to its
from healthy donors in culture and in vivo after transplanta- severity.34
tion.4,21 Individuals with aplastic anemia have elevated serum The antigens responsible for triggering and sustaining the
levels of erythropoietin, thrombopoietin, granulocyte colony- autoimmune attack on the stem cells are unknown. Possible
stimulating factor (G-CSF), and granulocyte-macrophage autoantigens have been identified using sera from aplastic
colony-stimulating factor (GM-CSF).22 In addition, the serum anemia patients; these include kinectin,35 diazepam-binding
level of FLT3 ligand, a growth factor that stimulates prolifera- inhibitor-related protein 1,36 and moesin.37 These proteins
tion of stem and progenitor cells, is up to 200 times higher are expressed in hematopoietic progenitor cells, but their
in patients with severe aplastic anemia than in healthy con- role in the pathogenesis of aplastic anemia requires further
trols.22,23 Despite their elevated levels, growth factors alone are investigation.
generally unsuccessful in correcting the cytopenias found in Approximately one third of patients with acquired aplastic
acquired aplastic anemia. anemia have shortened telomeres in their peripheral blood
The severe depletion of hematopoietic stem and progenitor granulocytes compared with aged-matched controls.38,39 Telo-
cells from the bone marrow may be due to (1) direct damage meres protect the ends of chromosomes from damage and
to stem cells, (2) immune damage to stem cells, or (3) other erosion, and cells with abnormally short telomeres undergo
unknown mechanisms. In the direct mechanism, a cytotoxic proliferation arrest and premature apoptosis. Approximately
drug, chemical, radiation, or virus damages the DNA of the 10% of patients with acquired aplastic anemia and shortened
stem and progenitor cells, causing apoptosis and cytolysis.4 telomeres have a mutation in the gene for either the ribonu-
In the immune mechanism, exposure to certain drugs, cleic acid (RNA) template (TERC) or the reverse transcriptase
chemicals, viruses, or other unknown agents causes an autoim- (TERT) of the telomerase complex.39-41 Telomerase is an
mune cytotoxic T lymphocyte attack that destroys the stem and enzyme complex that repairs and maintains the telomeres.
progenitor cells.24 An autoimmune pathophysiology was first The reason for the shortened telomeres in the other 90% of
suggested in the 1970s when aplastic anemia patients under patients is unknown, but may be the stressed hematopoiesis
going immunosuppressive conditioning before bone marrow brought on by the autoimmune attack or other unknown
transplantation experienced an improvement in cell counts.25 mutations.39 In stressed hematopoiesis, there is an increase in
Evidence supporting an autoimmune pathophysiology is that progenitor cell turnover, and the telomeres become shorter
in immune-related acquired aplastic anemia (1) there is an with each cell division. About 4% of patients with acquired
increase in cytotoxic (CD8+) T lymphocytes in the blood and aplastic anemia and short telomeres have mutations in the
bone marrow with an oligoclonal expansion of specific T-cell SBDS gene.42 The SBDS gene product is involved in ribosome
clones26; (2) these T cells produce increased amounts of cyto- biogenesis, but its relationship to telomere maintenance
kines, such as interferon- (IFN-) and tumor necrosis factor- is unknown.42 TERT/TERC and SBDS mutations occur in
(TNF-), that inhibit hematopoiesis and induce apoptosis27-29; the inherited aplastic anemias dyskeratosis congenita and
they also have an upregulation of T-bet, a transcription factor Shwachman-Diamond syndrome, respectively, and some
that binds to the promoter of the IFN- gene30; (3) there is an acquired aplastic anemia patients with these mutations may
increase in TNF- receptors on CD34+ cells31; and (4) there is actually have dyskeratosis congenita or Shwachman-Diamond
improvement in the cytopenia after immunosuppressive syndrome with an atypical presentation.3,4 Identification of
therapy (IST).4,24 Approximately two thirds of patients with these genetic defects in patients with acquired aplastic anemia
acquired aplastic anemia respond to IST; the one third who do has important implications for treatment in that IST may
not respond may have a severely depleted stem cell compart- not be effective and apparently healthy HLA-matched siblings
ment or other immune or nonimmune pathophysiologic who test positive for the same genetic mutation should
factors may be present.32 not be selected as donors for bone marrow transplantation.39
Possible mechanisms by which a drug, chemical, or virus Shortened telomeres occur more often in patients whose
may elicit an autoimmune response include mutation or alter- pancytopenia does not respond to IST,43 and a defect in
ation of stem cell antigens and disruption of immune regula- telomere maintenance may be another pathophysiologic
tion mechanisms. Concerning the former mechanism, a drug, mechanism of stem cell depletion, imparting a susceptibility
chemical, or virus may alter self-proteins, induce expression of to bone marrow failure after an environmental insult with a
abnormal or novel antigens, expose hidden antigens, or induce virus, drug, or autoimmune attack.39,41
CHAPTER 21 Bone Marrow Failure 287
Clinical Findings Blasts and other immature blood cells are characteristically
Symptoms vary in acquired aplastic anemia; the disorder can absent.
range from very severe to mild or asymptomatic. Patients Serum iron level and percent transferrin saturation are
usually present with symptoms typical of insidious-onset increased, which reflects the decreased use of iron for erythro-
anemia: pallor, fatigue, and weakness. Severe anemia can result poiesis. Liver function test results may be abnormal if the
in serious cardiac complications, including cardiac failure and pancytopenia was preceded by hepatitis. When high-resolution
death. Petechiae, bruising, epistaxis, bleeding gums, menor- flow cytometry is used, about two thirds of patients are found
rhagia, retinal hemorrhages, intestinal bleeding, and sometimes to have small numbers of PNH granulocytes (less than 25%)
intracranial bleeding may occur secondary to thrombocytope- in the peripheral blood,46 but only about 10% of patients
nia. Fever and bacterial or fungal infections are unusual at develop a large enough clone of PNH cells to display the
initial presentation but may occur after prolonged periods of typical clinical and biochemical manifestations of PNH.16 PNH
neutropenia. Splenomegaly and hepatomegaly are absent. is characterized by an acquired stem cell mutation that results
in circulating blood cells that lack glycosylphosphatidylinosi-
Laboratory Findings tol (GPI)linked proteins, such as CD55 and CD59. The
Pancytopenia is typical, although initially only one or two cell absence of CD55 and CD59 on the surface of the RBCs renders
lines may be decreased. The absolute neutrophil count is them more susceptible to lysis by complement. The signifi-
decreased, and the absolute lymphocyte count may be normal cance of the circulating PNH cells in aplastic anemia is uncer-
or decreased. The hemoglobin is usually less than 10g/dL, the tain, but it may be related to the ability of the GPI-deficient
mean cell volume (MCV) is increased or normal, and the stem cells to escape immune destruction.24,32 It is important to
percent and absolute reticulocyte counts are decreased. Table test for PNH cells in acquired aplastic anemia because of the
21-1 lists the diagnostic criteria for aplastic anemia by degree hemolytic and/or thrombotic complications associated with
of severity.6,8,44,45 This classification is helpful in guiding treat- PNH (see Chapter 23). The Ham test for complement-mediated
ment decisions. hemolysis is positive only if there are large numbers of circulat-
In the peripheral blood, neutrophils, monocytes, and plate- ing PNH cells; however, flow cytometry is a more sensitive
lets are decreased, and the RBCs are macrocytic or normocytic method and has replaced the Ham test for diagnosis of PNH
(Figure 21-1). Toxic granulation may be observed in the neu- (see Chapter 23).8,46,47
trophils, but the RBCs and platelets are normal in appearance. Bone marrow aspirates and biopsy specimens have promi-
nent fat cells with areas of patchy cellularity. Biopsy samples
are required for an accurate quantitative assessment of the
marrow cellularity, and severe hypocellularity is a characteristic
feature (Figure 21-2). Erythroid, granulocytic, and megakaryo-
cytic cells are decreased or absent. Dyserythropoiesis may be
present, but there is no dysplasia of the granulocyte or platelet
cell lines. Blasts and abnormal cell infiltrates are characteristi-
cally absent. Reticulin staining is normal.
In patients receiving IST, the risk of developing an abnormal
karyotype is 14% at 5 years and 20% at 10 years.48 Monosomy
7 and trisomy 8 are the most frequently identified cytogenetic
abnormalities.47,48 Cytogenetic analysis using conventional
culture techniques underestimates the incidence of karyotype
abnormalities because of the hypocellularity of the bone
marrow and scarcity of cells in metaphase.49 Interphase fluo-
Figure 21-1 Peripheral blood film for a patient with aplastic anemia rescent in situ hybridization (FISH) using deoxyribonucleic
(500). Note occasional macrocytes and absence of white blood cells and acid (DNA) probes for specific chromosome abnormalities has
platelets. the advantage of greater sensitivity and the ability to use

TABLE 21-1 Diagnostic Criteria for Aplastic Anemia


NSAA SAA VSAA
Bone marrow Hypocellular bone marrow plus at least two Bone marrow cellularity <25%* plus at least two of Same as SAA
of the following: the following:
Neutrophils ( 109/L) 0.5-1.5 0.2-0.5 <0.2
Platelets ( 109/L) 20-50 <20 Same as SAA
Other Hb 10 g/dL plus reticulocytes <30 109/L Reticulocytes <20 109/L or <1% corrected for Hct Same as SAA

Hb, Hemoglobin; Hct, hematocrit; NSAA, nonsevere aplastic anemia; SAA, severe aplastic anemia; VSAA, very severe aplastic anemia.
*Or 25% to 50% cellularity with <30% residual hematopoietic cells.
288 PART IV Hematopathology: Erythrocyte Disorders

HLA-identical sibling.4,8 Unfortunately, only 20% to 30% of


patients meet these criteria.4 For patients older than age 40
and those of any age without an HLA-identical sibling, IST,
consisting of antithymocyte globulin with cyclosporine, is the
preferred therapy.8,16 The antithymocyte globulin decreases
the number of activated T cells, and the cyclosporine inhibits
their function, suppressing the autoimmune reaction against
the stem cells. Approximately two thirds of patients initially
respond to IST but 30% to 40% eventually experience
relapse.16,32 For patients with severe acquired aplastic anemia
who are not responsive to IST, a second course of IST or a bone
marrow transplant from an HLA-matched unrelated donor are
options, but survival is not as high as with a bone marrow
A
transplant from an HLA-identical sibling.8,50 The response rate
for a second course of IST is approximately 65% for those who
experienced relapse and 30% for those whose disorder was
initially refractive to IST.51 Individuals with PNH cells (CD55
CD59) are almost twice as likely to respond to IST than are
those who lack these cells.46 In addition, the presence of both
PNH cells and HLA-DR2 increases the likelihood of response
by 3.5-fold.52 The use of G-CSF and other hematopoietic
growth factors as an adjunct to IST does not increase overall
survival nor improve the response rate; therefore, they are not
recommended for routine use.53,54
Other supportive therapy includes antibiotic and anti
fungal prophylaxis in cases of prolonged neutropenia. Patients
B with nonsevere aplastic anemia may not require treatment
but must be monitored periodically for cytopenia and abnor-
Figure 21-2 A, Normal bone marrow tissue section (hematoxylin and mal cells.
eosin stain). B, Hypoplastic bone marrow tissue section from a patient with
The overall outcome for patients with acquired aplastic
aplastic anemia (hematoxylin and eosin stain). (Courtesy Ann Bell, University
anemia has dramatically improved in the last two decades.
of Tennessee, Memphis.)
Among patients who receive a bone marrow transplant from
an HLA-identical sibling, 91% of children and 74% of adults
nondividing cells.49 In one study, FISH detected monosomy 7 achieve 10-year survival.50 Those percentages decrease to 75%
or trisomy 8 in 26% of aplastic anemia patients who had a of children and 63% of adults when the bone marrow trans-
normal karyotype by conventional cytogenetic testing.49 plant is from an HLA-matched unrelated donor.50 In patients
Patients with inherited aplastic anemia may be diagnosed treated with IST, 75% of children and 63% of adults achieve
erroneously as having acquired aplastic anemia if the disease 10-year survival.50 In addition, the 10-year risk of developing
first becomes apparent in late adolescence or adulthood or if hemolytic or thrombotic PNH is approximately 10%, and the
they lack the typical clinical and physical manifestations.3,4 risk of developing MDS or leukemia is 10% to 20% in the IST-
Exclusion of inherited aplastic anemias in the differential diag- treated group.16,47 Development of monosomy 7 predicts a
nosis of acquired aplastic anemia is essential, because these poor outcome with a greater likelihood of nonresponsiveness
conditions require a different therapeutical approach. The to IST and a progression to myelodysplastic syndrome (MDS)
inherited aplastic anemias are discussed later in the chapter. or leukemia.48

Treatment and Prognosis Inherited Aplastic Anemia


Severe aplastic anemia requires immediate treatment to prevent Patients with inherited aplastic anemia usually show manifes-
the dire consequences of serious pancytopenia. If a potential tations of the disorder at an early age and may have physical
causative agent is suspected, its use should be discontinued. To malformations. The three inherited diseases for which bone
keep blood counts at safe levels, platelets generally are admini marrow failure and pancytopenia are a consistent feature are
stered when the platelet count decreases to less than 10 109/L, Fanconi anemia, dyskeratosis congenita, and Shwachman-
whereas RBCs are given when the patient becomes symp Diamond syndrome.
tomatic.8 One of the most important early decisions that
must be made is whether a patient is a candidate for bone Fanconi Anemia
marrow transplantation. Bone marrow transplantation is Fanconi anemia (FA) is a chromosome instability disorder
generally the treatment of choice for patients with severe characterized by aplastic anemia, physical abnormalities, and
aplastic anemia who are younger than age 40 and have an cancer susceptibility. It was first described by Fanconi in 1927
CHAPTER 21 Bone Marrow Failure 289

in three brothers with skin pigmentation, short stature, and level is also increased.58 Chromosomes show abnormal fra
hypogonadism.55 FA occurs at a rate of 1 per 33,000. The carrier gility in FA. The addition of a DNA cross-linking agent such
rate is 1 in 300 in the United States and Europe, with a three- as diepoxybutane (DEB) or mitomycin C (MMC) to cultured
fold higher incidence in Ashkenazi Jews and South African peripheral blood lymphocytes causes characteristic chromo-
Africaners.56 FA is the most common of the inherited aplastic some breakage and is the diagnostic test for this disorder.3,58
anemias. Results of this test may be negative in the 10% to 15% of FA
patients who have somatic mosaicism due to a reversion of one
Clinical Findings. There is no typical presentation for FA. abnormal allele to the normal type.3,58 To confirm the diagnosis
Physical malformations may be present at birth or hematologic in these cases, the DEB chromosome breakage test or a flow
abnormalities may first appear during childhood or adulthood. cytometrybased MMC sensitivity test can be performed on
Only about two thirds of patients have physical malforma- cultured skin fibroblasts from a skin biopsy specimen.58,61 Lym-
tions.3,57 These anomalies vary considerably, although there is phocytes and skin fibroblasts can also be tested for FANCD2
a higher frequency of skeletal abnormalities (malformations of by immunoblotting after exposure to MMC, and a positive test
the thumbs, radial hypoplasia, microcephaly, hip dislocation, result is also diagnostic for FA.58
and scoliosis); skin pigmentation (hyperpigmentation or
hypopigmentation and caf-au-lait spots); short stature; and Treatment and Prognosis. By age 40, more than 90% of
abnormalities of the eyes, kidneys, and genitals.56-58 Low birth FA patients develop bone marrow failure; one third develop a
weight and developmental delays also are common. hematologic malignancy, particularly acute myeloid leukemia
The symptoms associated with pancytopenia usually (AML) and MDS; and one fourth develop other cancers.62 The
become apparent at 5 to 10 years of age; however, some median ages for development of leukemia and solid tumors
patients may be asymptomatic until adulthood.3,56 Individuals are 14 and 26 years, respectively.59 Squamous cell carcinomas
with FA have an increased risk of developing solid tumors, of the head and neck, anogenital region, and skin are the most
particularly oral, esophageal, anogenital, and hepatic tumors. common, followed by tumors of the liver, brain, and kidney.59
In approximately 5% of cases, a malignancy is diagnosed Compared with the general population, patients with FA have
before the FA is recognized.59 a risk of developing vulvar cancer, esophageal cancer, AML, and
head or neck cancer that is higher by 4300-fold, 2300-fold,
Genetics and Pathophysiology. FA is caused by biallelic 800-fold, and 700-fold, respectively.63 Approximately 3%
mutations or deletions in one of 13 genes: FANCA, FANCB, develop more than one type of cancer.62 Death by age 20 as a
FANCC, FANCD1 (also called BRCA2), FANCD2, FANCE, result of complications of bone marrow failure or malignancy
FANCF, FANCG (also called XRCC9), FANCI, FANCJ (also is common. Patients with mutations in the FANCC gene experi-
called BRIP1/BACH1), FANCL, FANCM, and FANCN (also ence bone marrow failure at an earlier age and have the poorest
called PALB2).3,57 The mode of inheritance is autosomal reces- survival.62 An increased rate of telomere shortening in FA cells
sive except for FANCB, which is X-linked. Mutations in the is associated with a greater severity of pancytopenia and higher
FANCA gene occur with the highest frequency.3,57 The relation- risk of developing malignancies; however, the precise role of
ship between mutations in the FA genes and disease pathology telomere shortening in the evolution of bone marrow failure
is not clear. In FA cells are highly susceptible to chromosome and cancer is unclear.64
breakage after exposure to DNA cross-linking agents and also Supportive treatment for cytopenia includes transfusions
have accelerated telomere shortening and apoptosis, a late and administration of androgens and cytokines (G-CSF and
S-phase cell cycle delay, hypersensitivity to oxidants, and cyto- GM-CSF).56,58 Currently the only curative treatment for cytope-
kine dysregulation.3,57,58,60 nia is a bone marrow transplant, preferably from a mutation-
The range of functions of the FA proteins is not completely negative, HLA-identical sibling. Because of the underlying
known, but these proteins do participate in a highly elaborate genetic defect and the bone marrow transplant conditioning
DNA damage response pathway. The FA pathway consists of a regimens, patients still have an increased risk of developing
nuclear core complex, ID complex, and effector proteins.57,60 secondary malignancies even after successful transplanta-
FA proteins A, B, C, E, F, G, L, and M form the nuclear core tion.58,62 Gene therapy has been attempted in clinical trials but
complex; proteins D2 and I form the ID complex; and the has not been successful.
effector proteins are D1, J and N.57,60 The core complex facili-
tates the monoubiquitylation and activation of the ID complex, Dyskeratosis Congenita
and the ID complex then localizes with effector DNA repair Dyskeratosis congenita (DC) is a rare inherited bone
proteins at foci of DNA damage to effect DNA repair.57,60 marrow failure syndrome with fewer than 600 known cases
worldwide.65
Laboratory Findings. Laboratory results are similar to
those in acquired aplastic anemia with pancytopenia, reticulo- Clinical Findings. DC is characterized by mucocutaneous
cytopenia, and a hypocellular bone marrow. Macrocytic RBCs abnormalities, bone marrow failure, and pancytopenia. The
are often the first detected abnormality, and thrombocytopenia typical clinical presentation includes abnormal skin pigmenta-
usually precedes the development of the other cytopenias.58 tion, dystrophic nails, and oral leukoplakia. The skin and nail
The Hb F level may be strikingly elevated, and the -fetoprotein manifestations usually appear before 10 years of age.3,58 The
290 PART IV Hematopathology: Erythrocyte Disorders

median age of diagnosis is 15 years, but DC can be diagnosed response in 50% to 70% of patients, it does not halt the pro-
at any age.66 By the age of 30 years, 80% to 90% of patients gression of the bone marrow failure.3
have bone marrow abnormalities.3 Patients with DC can also
manifest a wide range of multisystem abnormalities, including Shwachman-Diamond Syndrome
pulmonary fibrosis, liver disease, developmental delay, short Shwachman-Diamond syndrome (SDS), also called Shwachman-
stature, microcephaly, prematurely gray hair and hair loss, Bodian-Diamond syndrome, is an inherited multisystem disorder
immunodeficiency, dental caries and periodontal disease.66 characterized by pancreatic insufficiency, cytopenia, skeletal
Patients also have a 40% risk of developing cancer by age 50, abnormalities, and a predisposition for hematologic malignan-
with AML, MDS, and epithelial malignancies most commonly cies. The incidence has been estimated at approximately 1
diagnosed.65 in 76,500.68

Genetics and Pathophysiology. The chromosomes of Clinical Findings. Patients with SDS have peripheral
patients with DC have very short telomeres, and inherited blood cytopenia and a decrease in pancreatic enzyme secre-
defects in the telomerase complex are implicated in the patho- tion.45 The pancreatic insufficiency causes symptoms of malab-
physiology.66 The telomerase complex synthesizes telomere sorption that usually appear in early infancy.3 Due to
repeats to elongate the ends of chromosomes, and maintaining neutropenia and immune dysfunction, patients are at risk of
the length of the telomeres is important for cell survival. Six severe infections, including fatal overwhelming sepsis.45,69
mutated genes and three inheritance patterns are recognized in Almost all SDS patients have delayed bone maturation and
DC: autosomal dominant, X-linked recessive, and autosomal approximately half have short stature.45
recessive.3,66 The autosomal dominant type is due to mutations
in genes that code for TERC (RNA component of telomerase), Genetics and Pathophysiology. SDS is an autosomal
TERT (telomerase enzyme), or TINF2 (component of the shel- recessive disorder, and 90% of patients have biallelic muta-
terin complex that regulates telomere length). The X-linked tions in the SBDS gene.3,45,69 The SBDS gene is involved in
recessive form is due to mutations in the gene that codes for ribosome metabolism and mitotic spindle stability,70,71 but its
the DKC1 protein or dyskerin. Dyskerin is a ribonucleoprotein relationship to the disease manifestations is unknown. There
involved in RNA processing, and it associates with TERC in the are quantitative and qualitative deficiencies in CD34+ cells,
telomerase complex. In the autosomal recessive type, muta- dysfunctional bone marrow stromal cells, increased apoptosis
tions in TERT, NHP2, and NOP10 have been identified.66 The and mitotic spindle destabilization in hematopoietic cells, and
proteins coded by the latter two genes are also involved in RNA short telomeres in peripheral blood granulocytes.3,45,69,71
processing and telomerase maintenance. Although the exact
pathophysiologic mechanisms are still unknown, the exces- Laboratory Findings. Nearly all SDS patients have neu-
sively short telomeres in DC cause premature death in the tropenia (less than 1.5 109 neutrophils/L) that is persistent
rapidly dividing cells in the bone marrow and epithelium, and or intermittent.72 Half of the patients develop anemia or
likely lead to genomic instability and a predisposition to thrombocytopenia, and one fourth develop pancytopenia.72
cancer.3,58,67 The RBCs are usually normocytic but can be macrocytic, and
approximately two thirds of patients have an increase in Hb
Laboratory Findings. Pancytopenia and macrocytic RBCs F.45,72 The bone marrow is usually hypocellular, but can be
are typical peripheral blood findings. The Hb F level may also normal or even hypercellular. Due to the pancreatic insuffi-
be increased. Only about 40% of patients have a mutation in ciency, the 72-hour fecal fat test shows increased excretion of
one of the six telomerase complex genes identified to date.67 A fat, and the serum trypsinogen and isoamylase levels are
new flow FISH test for detection of very short telomeres in decreased compared with the age-related reference ranges,69
WBC subsets (including total lymphocytes, naive T cells, and but the sweat chloride test is normal.
B cells) has been proposed as a diagnostic test for those with
symptoms or a family history of DC who lack mutations in Treatment and Prognosis. Treatment consists of G-CSF
known genes.67 Patients with FA, Shwachman-Diamond syn- for the neutropenia, RBC and platelet transfusions for the
drome, and acquired aplastic anemia may also have cells with anemia and thrombocytopenia, and pancreatic enzyme replace-
short telomeres; however, in contrast to DC, the short telo- ment for the pancreatic insufficiency. The risk of AML and MDS
meres are not found in multiple WBC subsets.67 is approximately 19% at 20 years and 36% at 30 years.73 Allo-
geneic bone marrow transplantation is recommended in cases
Treatment and Prognosis. Approximately 60% to 70% of severe pancytopenia, AML, or MDS, but overall survival is
of deaths in patients with DC are due to the effects of bone only about 60% to 65%.45,69
marrow failure, 10% to 15% develop fatal pulmonary disease,
and another 10% die because of complications of malignan- Differential Diagnosis
cies.3,58 The median survival is 42 years.65 Treatment with bone A differential diagnosis must be made between acquired aplas-
marrow transplantation has not been optimal because of the tic anemia, inherited aplastic anemia, and other causes of pan-
high incidence of fatal pulmonary fibrosis and vascular comp cytopenia, including PNH, MDS, megaloblastic anemia, acute
lications.3,66 Although androgen therapy produces a transient leukemia, and hairy cell leukemia. The key features that
CHAPTER 21 Bone Marrow Failure 291

TABLE 21-2 Differentiation of Aplastic Anemia from Other Causes of Pancytopenia


Condition Peripheral Blood Bone Marrow Laboratory Test Results Clinical Findings
Failure of Bone Marrow to Produce Blood Cells
Aplastic anemia No immature WBCs or RBCs; Hypocellular; blasts and abnormal Acquired: PNH cells* may be Splenomegaly absent
reticulocytes; MCV or cells absent; reticulin normal; RBC present; chromosome
normal dyspoiesis may be present; WBC abnormalities may be present
and platelet dyspoiesis absent Inherited: see Table 21-3

Increased Destruction of Blood Cells


PNH Reticulocytes ; MCV normal Erythroid hyperplasia; may be PNH cells* present; Splenomegaly absent;
or ; nucleated RBCs hypocellular hemoglobinuria +/; thrombosis may be
present or absent chromosome abnormalities present
may be present

Ineffective Hematopoiesis
Myelodysplastic Variable pancytopenia; Hypercellular; 20% of cases Chromosome abnormalities Splenomegaly
syndrome reticulocytes ; MCV hypocellular; dyspoiesis in one or usually present uncommon
normal or ; blasts and more cell lines present; blasts and
abnormal WBCs, RBCs, and immature cells present; reticulin
platelets may be present
Megaloblastic MCV ; oval macrocytes; Hypercellular with megaloblastic Serum vitamin B12 or folate or Splenomegaly absent
anemias hypersegmented neutrophils features both

Bone Marrow Infiltration


Acute leukemia Blasts present Hypercellular; blasts ; reticulin Chromosome abnormalities may Splenomegaly may be
be present present
Hairy cell Hairy cells present; Hairy cells and fibrosis present; Hairy cells present; TRAP + Splenomegaly present
leukemia monocytes reticulin (60-70% of cases)

, Increased; , decreased; +, positive result; +/, positive or negative result; MCV, mean cell volume; PNH, paroxysmal nocturnal hemoglobinuria; RBC, red blood cell;
TRAP, tartrate-resistant acid phosphatase; WBC, white blood cell.
*PNH cells are detected by flow cytometry by their lack of expression of CD55 and CD59.

Hairy cells are detected by flow cytometry by their expression of CD20, CD11c, CD25, and FMC7.

distinguish these conditions are listed in Table 21-2 and Table normal WBC and platelet counts. PRCA may be acquired or
21-3.3,8 These conditions require different therapeutic regi- congenital. It is important to distinguish between the acquired
mens; therefore, it is essential to distinguish them so that the and congenital forms, because they require different therapeu-
patient receives the appropriate treatment. In addition, recog- tic approaches.
nition of PNH is important because of the hemolysis and
thrombosis associated with the disease (see Chapter 23). Acquired Pure Red Cell Aplasia
Lymphomas, Hodgkin disease, myelofibrosis, and myco- Acquired PRCA may occur in children or adults and can be
bacterial infections occasionally may present with pancytope- acute or chronic. Primary PRCA may be idiopathic or auto
nia but can be distinguished through careful examination of immune related. Secondary PRCA may occur in association
the bone marrow and, in the case of lymphomas, molecular with an underlying thymoma, hematologic malignancy, solid
tests for chromosome abnormalities. Anorexia nervosa also tumor, infection, chronic hemolytic anemia, collagen vascular
may present with pancytopenia, but the bone marrow is hypo- disease, or exposure to drugs or chemicals.74,75 Therapy is first
cellular and has a decrease number of fat cells.8 directed at treatment of the underlying condition, but if the
PRCA is not responsive, IST is instituted. Cyclosporine is asso-
ciated with a higher response rate (65% to 87%) than cortico-
OTHER FORMS OF BONE
steroids (30% to 62%), and it is better suited for long-term
MARROW FAILURE
maintenance if needed.75
Pure Red Cell Aplasia The acquired form of PRCA in young children is also known
Pure red cell aplasia (PRCA) is a rare disorder of erythropoiesis as transient erythroblastopenia of childhood (TEC). A history of
characterized by a selective and severe decrease in erythrocyte viral infection is found in half of patients and an immune
precursors in an otherwise normal bone marrow. Patients have mechanism is involved in its etiology. The anemia is normo-
severe anemia (usually normocytic), reticulocytopenia, and cytic, and Hb F and erythrocyte adenosine deaminase levels are
292 PART IV Hematopathology: Erythrocyte Disorders

TABLE 21-3 Key Characteristics of Inherited/Congenital Bone Marrow Failure Anemias


Condition Genetics* Peripheral Blood Bone Marrow Laboratory Test Results Clinical Findings
Due to Bone Marrow Hypoplasia
FA AR, XLR Pancytopenia; Hypocellular with in all cell Chromosome breakage with DEB/ Physical malformations may
13 genes reticulocytes ; lines MMC; Hb F may be be present; risk of
MCV cancers, leukemia,
myelodysplastic syndrome
DC AR, XLR, AD Pancytopenia; Hypocellular with in all cell 40% have mutations in TERC, Physical malformations may
6 genes reticulocytes ; lines TERT, DKC1, TINF2, NHP2, or be present; pulmonary
MCV NOP10; Hb F may be ; very disease; risk of cancers,
short telomeres in lymphocyte leukemia, myelodysplastic
subsets syndrome
SDS AR Neutropenia; Hypocellular, normocellular, 90% have mutations in SBDS Pancreatic insufficiency;
1 gene pancytopenia or hypercellular gene; serum trypsinogen and physical malformations
(25% of cases); isoamylase for age; Hb F may be present; risk of
reticulocytes ; may be infections, leukemia,
MCV normal or myelodysplastic syndrome
DBA AD (50% of Anemia; reticulocytes Erythroid hypoplasia Erythrocyte adenosine deaminase Physical malformations may
cases) ; MCV ; Hb F may be ; 25% have be present; risk of
6 genes mutations in RPS19 gene; cancers, leukemia,
another 25% have mutations myelodysplastic syndrome
in RPS17, RPS24, RPL5,
RPL11, or RPL35A
Due to Ineffective Hematopoiesis
CDA I AR Anemia; reticulocytes Hypercellular; RBC precursors Mutations in CDAN1 gene; Physical malformations may
1 gene ; MCV ; poik, megaloblastoid with spongy, Swiss cheese be present; iron overload;
baso stipp, Cabot internuclear chromatin heterochromatin in splenomegaly;
rings bridges and <5% erythroblasts by electron hepatomegaly
binucleated forms microscopy
CDA II AR Anemia; reticulocytes Hypercellular; RBC precursors Positive Ham test result; Physical malformations may
1 gene ; MCV normal; normoblastic with 10-35% mutations in SEC23B gene be present; iron overload;
poik, baso stipp binucleated forms jaundice; gallstones;
splenomegaly
CDA III AD Mild anemia; Hypercellular; RBC precursors None Treatment usually not
Gene unknown reticulocytes ; megaloblastoid with giant needed
MCV ; poik, multinucleated forms with
baso stipp up to 12 nuclei

, Increased; , decreased; AR, autosomal recessive; AD, autosomal dominant; baso stipp, basophilic stippling; CDA, congenital dyserythropoietic anemia; DBA, Diamond-Blackfan
anemia; DC, dyskeratosis congenita; DEB, diepoxybutane; FA, Fanconi anemia; Hb, hemoglobin; MCV, mean cell volume; MMC, mitomycin C; poik, poikilocytosis; RBC, red blood
cell; SDS, Shwachman-Diamond syndrome; SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel electrophoresis; XLR, X-linked recessive.
*Genes identified as of 2009; genetic discovery is ongoing.

normal.74,76 Transfusions are the initial therapy, and restoration Mutations in these ribosomal proteins disrupt ribosome bio-
of normal erythropoiesis occurs in most patients. There may genesis in DBA, but the pathophysiologic mechanisms leading
be a genetic predisposition to TEC in some families.76 to the clinical manifestations are unknown. Almost half of
DBA cases are linked to an autosomal dominant inheritance
Congenital Pure Red Cell Aplasia: pattern, but sporadic mutations are also found.77
Diamond-Blackfan Anemia Over 90% of patients show signs of the disorder during
Diamond-Blackfan anemia (DBA) is a congenital erythroid the first year of life at a median age of 8 weeks; however,
hypoplastic disorder of early infancy with an estimated occur- some patients with DBA are asymptomatic until adulthood.78
rence rate of 1 in 143,000.58,77 In DBA mutations have been Approximately half of patients have physical anomalies such
identified in six genes that code for structural ribosome pro- as craniofacial dysmorphism, short stature, and neck and
teins: RPS19, RPS17, and RPS24 in the 40S subunit and RPL5, thumb malformations.58,78
RPL11, and RPL35A in the 60S subunit.77 Approximately 25% The characteristic peripheral blood finding is a severe mac-
of patients have a mutation in the RPS19 gene, and mutations rocytic anemia with reticulocytopenia.58 The WBC count is
in the other five genes account for another 25% of cases.77 normal or slightly decreased, and the platelet count is normal
CHAPTER 21 Bone Marrow Failure 293
or slightly increased. Bone marrow examination distinguishes
DBA from the hypocellular marrow in aplastic anemia, because
there is normal cellularity of myeloid cells and megakaryocytes
and hypoplasia of erythroid cells. The karyotype in DBA is
normal. In most cases levels of Hb F and erythrocyte adenosine
deaminase are increased; these findings distinguish it from
TEC, in which these levels are normal.58,77 Therapy includes
RBC transfusions and corticosteroids. Although 50% to 75%
of patients respond to corticosteroid therapy, the side effects
are severe with long-term use.58,77 Overall survival is 75% at 40
years.78 After treatment with bone marrow transplantation, the
survival rate is over 90% in patients under 10 years of age with
a mutation-negative, HLA-matched related donor and 80% in
those with an unrelated donor.77
Figure 21-3 Erythrocyte precursors with nuclear bridging indicating dys-
Congenital Dyserythropoietic Anemia erythropoiesis (bone marrow, 1000). (Modified from Carr JH, Rodak BF:
The congenital dyserythropoietic anemias (CDAs) are a hetero- Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
geneous group of rare disorders characterized by refractory
anemia, reticulocytopenia, hypercellular bone marrow with
markedly ineffective erythropoiesis, and distinctive dysplastic invaginated, carrying cytoplasmic organelles into the nucleus.
changes in bone marrow erythroblasts. Megaloblastoid devel- Treatment includes administration of interferon- and iron
opment occurs in some types, but it is not related to vitamin depletion.58,80
B12 or folate deficiency. Granulopoiesis and thrombopoiesis CDA II is the most common type and is autosomal reces-
are normal. The anemia varies from mild to moderate, even sive. More than 300 cases have been reported.80 It is caused by
among affected siblings. Secondary hemosiderosis arises from mutations in the SEC23B gene on chromosome 20.83 SEC23B
the chronic intramedullary and extramedullary hemolysis of encodes a component of the coat protein complex (COPII)
erythroblasts and circulating RBCs and the increase in iron that forms vesicles for transport of secretory proteins from
absorption associated with the accelerated inefficient erythro- the endoplasmic reticulum to the Golgi apparatus,84 but its
poiesis. Iron overload develops even in the absence of role in the pathophysiology of CDA II is unknown. The anemia
blood transfusions. Jaundice and cholelithiasis (as a result of in CDA II is mild to moderate; hemoglobin ranges from 9 to
hyperbilirubinemia) and splenomegaly are common findings. 12g/dL, with a mean of 11g/dL.79 RBCs are usually nor
CDAs do not progress to aplastic anemia or hematologic mocytic with anisocytosis, poikilocytosis, and basophilic
malignancies.79 stippling. The bone marrow has normoblastic erythropoiesis
Symptoms of CDA usually occur in childhood or adoles- with 10% to 35% binucleated forms and rare multinucleated
cence but may first appear in adulthood.58 CDA is classified forms; occasional pseudo-Gaucher cells are also seen.79 Circu-
into three major types: CDA I, CDA II, and CDA III. There lating RBCs test positive on the Ham acidified serum test (but
are rare variants that do not fall into these categories, and not on the sucrose hemolysis test); for this reason, CDA II is
they have been assigned to four other groups, numbered IV also known as HEMPAS (hereditary erythroblastic multinucle-
through VII.58,79 arity with positive acidified serum).58 The Ham test is no longer
CDA I is autosomal recessive and is characterized by a mild used for confirmation of CDA II because of the difficulty in
to severe chronic anemia. Over 150 cases of CDA I have been performing the appropriate quality control and the lack of
reported.80 It is caused by mutations in the CDAN1 gene on availability of the test in many laboratories.79 RBCs also agglu-
chromosome 15.81 CDAN1 codes for codanin-1, a cell cycle tinate with anti-i antisera and show abnormal migration of
regulated nuclear protein,82 but its role in the pathophysiology band 3 using sodium dodecyl sulfate polyacrylamide gel
of CDA I is unknown. Malformations of fingers or toes, brown electrophoresis.79 Treatment includes splenectomy and iron
skin pigmentation, and neurologic defects are found more depletion.58,80
frequently in CDA I than in the other types. The hemoglobin CDA III is the least common of the CDAs. A familial auto-
is usually in the range of 6.5 to 11.5g/dL, with a mean of somal dominant form has been found in a small number of
9.5g/dL.79 RBCs are macrocytic and may exhibit marked families and is mapped to chromosome 15; the nonfamilial or
poikilocytosis, basophilic stippling, and Cabot rings. The sporadic form is extremely rare, with fewer than 20 cases
erythroblasts are megaloblastoid and characteristically have reported.80 The anemia is mild, and the hemoglobin is usually
internuclear chromatin bridges or nuclear strands between two in the range of 8 to 14g/dL, with a mean of 12g/dL.79 RBCs
erythroblasts or between two nuclei in a single cell (Figure are generally macrocytic with poikilocytosis and basophilic
21-3). There are less than 5% binucleated erythroblasts. Ultra- stippling. The bone marrow has megaloblastic development,
structurally, the characteristic feature of the CDA I erythroblast and giant erythroblasts with up to 12 nuclei are a characteristic
is a spongy heterochromatin with a Swiss cheese appear- feature. Patients are transfusion independent, and clinical iron
ance.79 In addition, nuclear membranes can be disrupted and overload is not observed.
294 PART IV Hematopathology: Erythrocyte Disorders

Myelophthisic Anemia gastrointestinal bleeding. In addition, chronic inflammation


Myelophthisic anemia is the infiltration of abnormal cells into and an iron-poor diet may limit the iron available for erythro-
the bone marrow and subsequent destruction and replacement poiesis.87,88 Anemia in CKD is usually normocytic and normo-
of the normal hematopoietic cells. Metastatic solid tumor cells chromic, with normal or decreased reticulocytes. In addition,
(particularly from lung, breast, and prostate), leukemic cells, burr cells may be present as a result of the uremia.87
fibroblasts, and inflammatory cells (found in miliary tubercu- Anemia in CKD can lead to cardiovascular complications,
losis and fungal infections) are implicated.85,86 Cytokines, faster progression to kidney failure, and a suboptimal quality of
growth factors, and other substances are released that suppress life.87 The Kidney Disease Outcomes Quality Initiative of the
hematopoiesis or destroy stem, progenitor, or stromal cells, National Kidney Foundation recommends an annual assess-
which results in peripheral cytopenias.86 If the infiltration ment of hemoglobin in patients with CKD and an investigation
and proliferation of the abnormal cells disrupts the normal of the anemia if the hemoglobin is less than 13.5g/dL in adult
bone marrow architecture, premature release of immature cells men and less than 12.0g/dL in adult women.87 Treatment of
from the bone marrow occurs. In addition, because of the anemia resulting from CKD involves the administration of
unfavorable bone marrow environment, stem and progenitor recombinant human erythropoietin or other erythropoiesis-
cells migrate to the spleen and liver and establish extra stimulating agents (ESAs) with the goal of keeping the hemo-
medullary hematopoietic sites.86 Because blood cell production globin generally between 11g/dL and 12g/dL.87,92 Maintaining
at these extramedullary sites is less efficient, immature cells the hemoglobin above 13g/dL is not recommended because of
also may be released prematurely into the circulation from an increase in the risk of cardiovascular and thromboembolitic
these organs.85 complications.87,92 Successful ESA therapy requires adequate
Myelophthisic anemia is typically mild to moderate with iron stores, so plasma ferritin level and percent transferrin
normocytic erythrocytes and reticulocytopenia. The typical saturation are also monitored. Iron is administered with ESA
findings in the peripheral blood are teardrop erythrocytes and therapy to maintain the transferrin saturation above 20% and
nucleated RBCs, but immature myeloid cells (leukoerythro- the plasma ferritin level above 100ng/mL for nondialysis-
blastic peripheral blood picture), megakaryocyte fragments, dependent CKD patients and above 200ng/mL for hemodialysis-
and giant platelets also may be present.85 Myelophthisic anemia dependent CKD patients.87 Iron therapy is not routinely
is distinguished from aplastic anemia by the presence of nor- recommended if the ferritin level is above 500ng/mL.87
mocytic RBCs with teardrop forms, a leukoerythroblastosis in Patients may become hyporesponsive to ESA therapy
the peripheral blood, and abnormal cells in the bone marrow because of functional iron deficiency (FID). In FID, the bone
aspiration or biopsy specimen. marrow is unable to release iron rapidly enough to accommo-
date the accelerated rate of erythropoiesis. The transferrin satu-
Anemia of Chronic Kidney Disease ration remains below 20%, but the serum ferritin level is
Anemia occurs in most patients with chronic kidney disease normal or increased, indicating adequate iron stores.93 Patients
(CKD), and its prevalence increases with the severity of the with FID are unable to reach or maintain the target hemoglo-
disease.87,88 In 1999 to 2004 in the United States, 13.1% of bin, even with high doses of ESAs; they will, however, be able
adults over 20 years of age had CKD, or approximately 26 to reach the target hemoglobin after intravenous iron therapy.93
million adults.89 The major cause of the anemia in CKD is the Methods that have been proposed for diagnosis of FID in
inadequate production of erythropoietin by the kidneys.87,88 kidney disease include a decrease in the reticulocyte hemoglo-
Without erythropoietin, the bone marrow is unable to increase bin content, an increase in soluble transferrin receptor level,
RBC production in response to tissue hypoxia, and anemia and the presence of greater than 10% hypochromic red blood
ensues. Other factors also contribute to the anemia in CKD. cells in the peripheral blood.93,94 Some other causes of hypore-
Uremic toxins accumulate in the blood because of the kidney sponsiveness to ESA therapy include chronic inflammatory
failure, and they inhibit erythropoiesis and shorten the life span disease, infection, malignancy, aplastic anemia, antibody-
of the RBCs.90,91 Patients experience chronic blood loss and iron mediated PRCA, thalassemia, multiple myeloma, and the pres-
deficiency due to hemodialysis, frequent blood draws, and ence of Hb H or Hb S.87

SUMMARY
Bone marrow failure is the reduction or cessation of blood cell exposures, radiation, viruses, or miscellaneous conditions such as
production affecting one or more cell lines. Pancytopenia, or a PNH, autoimmune diseases, and pregnancy. Inherited aplastic
decrease in RBCs, WBCs, and platelets, is a common finding. anemias includes FA, DC, and SDS.
Sequelae of pancytopenia include weakness and fatigue, infec- Bone marrow failure in acquired aplastic anemia is due to the
tions, and bleeding. destruction of hematopoietic stem cells either by the direct toxic
Aplastic anemia may be acquired or inherited. Acquired aplastic effects of a drug, by an autoimmune T-cell attack against the
anemia may be idiopathic or secondary to drugs, chemical stem cells, or by other unknown mechanisms. The autoimmune
CHAPTER 21 Bone Marrow Failure 295

reactions are a rare adverse event after exposure to a drug, chemi- Defects in the telomerase complex play a role in the pathophysio
cal, or virus and are called idiosyncratic in that they are unpredict- logy of inherited aplastic anemia and some types of acquired
able and unrelated to the dose or duration of exposure. aplastic anemia by causing the inability to elongate telomeres at
Aplastic anemia is classified as nonsevere, severe, or very severe the ends of chromosomes; this leads to premature stem cell
based on bone marrow hypocellularity, absolute neutrophil count, senescence and death.
platelet count, hemoglobin level, and reticulocyte count. The PRCA affects only erythropoiesis. TEC is the acquired form, and
severity classification helps to guide treatment decisions. DBA is the inherited form. DBA usually manifests in early infancy,
Treatment for severe and very severe acquired aplastic anemia is and mutations have been identified in six genes that code for
bone marrow transplantation in younger patients with an HLA- ribosomal proteins.
identical sibling or IST with antithymocyte globulin and cyclospo- Patients with CDA exhibit refractory anemia, reticulocytopenia,
rine for patients who are not candidates for transplantation. secondary hemosiderosis, and distinct abnormalities of erythroid
FA, DC, and SDS are inherited forms of aplastic anemia with precursors. Three major types are recognized: CDA I, CDA II, and
progressive bone marrow failure and pancytopenia that may CDA III.
present with or without physical malformations. FA is autosomal Myelophthisic anemia results from the replacement of normal
recessive or X-linked, and mutations in 13 genes have been bone marrow with abnormal cells. The anemia of CKD is mainly
identified. Chromosome breakage when DEB or MMC is added to due to the inadequate production of erythropoietin by the kidneys.
cultured peripheral blood lymphocytes is diagnostic. DC can be
X-linked, autosomal dominant, or autosomal recessive, and muta- Now that you have completed this chapter, go back and
tions in six genes have been identified. SDS is autosomal reces- read again the case study at the beginning and respond
sive, and mutations in one gene have been identified. to the questions presented.

R E V I E W Q UESTIONS
1. The clinical consequences of pancytopenia include: 5. The most consistent peripheral blood findings in severe
a. Pallor and thrombosis aplastic anemia are:
b. Kidney failure and fever a. Hairy cells, monocytopenia, and neutropenia
c. Fatigue, infection, and bleeding b. Macrocytosis, thrombocytopenia, and neutropenia
d. Weakness, hemolysis, and infection c. Blasts, immature granulocytes, and thrombocytopenia
d. Polychromasia, nucleated RBCs, and hypersegmented
2. Idiopathic acquired aplastic anemia is due to a(n): neutrophils
a. Drug reaction
b. Benzene exposure 6. The treatment that has shown the best success rate in
c. Inherited mutation in stem cells young patients with severe aplastic anemia is:
d. Unknown cause a. IST
b. Long-term RBC and platelet transfusions
3. The pathophysiologic mechanism in acquired idiosyn- c. Administration of hematopoietic growth factors and
cratic aplastic anemia is: androgens
a. Replacement of bone marrow by abnormal cells d. Stem cell transplant from an HLA-identical sibling
b. Destruction of stem cells by autoimmune T cells
c. Defective production of hematopoietic growth 7. The test that is most useful in differentiating FA from other
factors causes of pancytopenia is:
d. Inability of bone marrow stroma to support stem a. Bone marrow biopsy
cells b. Ham acidified serum test
c. DEB-induced chromosome breakage
4. Based on the criteria in Table 21-1, what is the aplastic d. Flow cytometric analysis of CD55 and CD59 cells
anemia classification of a patient with a bone marrow
cellularity of 10%, an Hb of 7g/dL, an absolute neu 8. Mutations in genes that code for the telomerase complex
trophil count of 0.1 109/L, and a platelet count of may induce bone marrow failure by causing which of the
10 109/L? following?
a. Nonsevere a. Resistance of stem cells to normal apoptosis
b. Moderate b. Autoimmune reaction against telomeres in stem cells
c. Severe c. Decreased production of hematopoietic growth factors
d. Very severe d. Premature death of stem cells
296 PART IV Hematopathology: Erythrocyte Disorders

9. DBA differs from inherited aplastic anemia in that in the 11. The primary pathophysiologic mechanism of anemia of
former: CKD is:
a. Reticulocyte count is increased a. Inadequate production of erythropoietin
b. Hb F level is decreased b. Excessive hemolysis
c. Only erythropoiesis is affected c. Stem cell mutation
d. Congenital malformations are absent d. Toxic destruction of stem cells

10. Which anemia should be suspected in a patient with


refractory anemia, reticulocytopenia, hemosiderosis, and
binucleated erythrocyte precursors in the bone marrow?
a. FA
b. DC
c. Acquired aplastic anemia
d. CDA

REFERENCES
1. Ehrlich P: ber einen Fall von Anmie mit Bemerkungen 16. Frickhofen N, Heimpel H, Kaltwasser JP, et al: Antithymocyte
ber regenerative Vernderungen des Knochenmarks. Charite globulin with or without cyclosporin A: 11-year follow-up of
Annal 13:301, 1888. a randomized trial comparing treatments of aplastic anemia.
2. Vaquez MH, Aubertin C: Lanmie pernicieuse daprs les Blood 101:1236-1242, 2003.
conceptions actuelles. Bull Soc Med Hop Paris 21:288-297, 17. Maciejewski JP, Selleri C, Sato T, et al: A severe and consistent
1904. deficit in marrow and circulating primitive hematopoietic
3. Dokal I, Vulliamy T: Inherited aplastic anaemias/bone cells (long-term culture-initiating cells) in acquired aplastic
marrow failure syndromes. Blood Rev 22:141-153, 2008. anemia. Blood 88:1983-1991, 1996.
4. Young NS, Maciejewski JP: Aplastic anemia. In Hoffman R, 18. Philpott NJ, Scopes J, Marsh JCW, et al: Increased apoptosis
Benz EJ, Shattil SJ, et al, editors: Hematology: basic principles in aplastic anemia bone marrow progenitor cells: possible
and practice, ed 5, Philadelphia, 2009, Churchill Livingstone, pathophysiologic significance. Exp Hematol 23:1642-1648,
pp 359-394. 1995.
5. Muir KR, Chilvers CED, Harriss C, et al: The role of occupa- 19. Maciejewski JP, Selleri C, Sato T, et al: Increased expression
tional and environmental exposures in the aetiology of of Fas antigen on bone marrow CD34 sup+ cells of patients
acquired severe aplastic anaemia: a case control investigation. with aplastic anaemia. Br J Haematol 91:245-252, 1995.
Br J Haematol 123:906-914, 2003. 20. Zeng W, Chen G, Kajigaya S, et al: Gene expression profiling
6. International Agranulocytosis and Aplastic Anemia Study: in CD34 cells to identify differences between aplastic
Incidence of aplastic anemia: the relevance of diagnostic cri- anemia patients and healthy volunteers. Blood 103:325-332,
teria. Blood 70:1718-1721, 1987. 2004.
7. Kojima S: Aplastic anemia in the Orient. Int J Hematol 21. Novitzky N, Jacobs P: Immunosuppressive therapy in bone
76(suppl 2):173-174, 2002. marrow aplasia: the stroma functions normally to support
8. Marsh JCW, Ball SE, Darbyshire P, et al: Guidelines for the hematopoiesis. Exp Hematol 23:1472-1477, 1995.
diagnosis and management of acquired aplastic anaemia. 22. Koijima S: Hematopoietic growth factors and marrow stroma
Br J Haematol 123:782-801, 2003. in aplastic anemia. Int J Hematol 68:19-28, 1998.
9. Nimer SD, Ireland P, Meshkinpour A, et al: An increased HLA 23. Wodnar-Filipowicz A, Lyman SD, Gratwohl A, et al: Flt3
DR2 frequency is seen in aplastic anemia patients. Blood ligand level reflects hematopoietic progenitor cell function in
84:923-927, 1994. aplastic anemia and chemotherapy-induced bone marrow
10. Saunthararajah Y, Nakamura R, Nam JM, et al: HLA-DR15 aplasia. Blood 88:4493-4499, 1996.
(DR2) is overrepresented in myelodysplastic syndrome and 24. Young NS, Maciejewski J: The pathophysiology of acquired
aplastic anemia and predicts a response to immunosuppres- aplastic anemia. N Engl J Med 336:1365-1372, 1997.
sion in myelodysplastic syndrome. Blood 100:1570-1574, 25. Mathe G, Amiel JL, Schwarzenberg L, et al: Bone marrow
2002. graft in man after conditioning by antilymphocytic serum.
11. Dougherty D, Garte S, Barchowsky A, et al: NQO1, MPO, Br Med J 2:131-136, 1970.
CYP2E1, GSTT1 and GSTM1 polymorphisms and biological 26. Risitano AM, Maciejewski JP, Green S, et al: In-vivo dominant
effects of benzene exposurea literature review. Toxicol Lett immune responses in aplastic anaemia: molecular tracking of
182:7-17, 2008. putatively pathogenic T-cell clones by TCR--CDR3 sequenc-
12. Sutton JF, Stacey M, Kearns WG, et al: Increased risk for aplas- ing. Lancet 364:355-364, 2004.
tic anemia and myelodysplastic syndrome in individuals 27. Hara T, Ando K, Tsurumi H, et al: Excessive production of
lacking glutathione S-transferase genes. Pediatr Blood Cancer tumor necrosis factor-alpha by bone marrow T lymphocytes
42:122-126, 2004. is essential in causing bone marrow failure in patients with
13. Brown KE, Tisdale J, Barrett AJ, et al: Hepatitis-associated aplastic anemia. Eur J Haematol 73:10-16, 2004.
aplastic anemia. N Engl J Med 336:1059-1064, 1997. 28. Young NS: Hematopoietic cell destruction by immune mech-
14. Choudhry VP, Gupta S, Gupta M, et al: Pregnancy associated anisms in acquired aplastic anemia. Semin Hematol 37:3-14,
aplastic anemiaa series of 10 cases with review of literature. 2000.
Hematol 7:233-238, 2002. 29. Selleri C, Sato T, Anderson S, et al: Interferon-gamma and
15. Stalder MP, Rov A, Halter J, et al: Aplastic anemia and con- tumor necrosis factor-alpha suppress both early and late
comitant autoimmune diseases. Ann Hematol 88:659-665, stages of hematopoiesis and induce programmed cell death.
2009. J Cell Physiol 165:538-546, 1995.
CHAPTER 21 Bone Marrow Failure 297
30. Solomou EE, Keyvanfar K, Young NS: T-bet, a Th1 transcrip- 51. Scheinberg P, Nunez O, Young NS: Retreatment with rabbit
tion factor, is up-regulated in T cells from patients with aplas- anti-thymocyte globulin and ciclosporin for patients with
tic anemia. Blood 107:3983-3991, 2006. relapsed or refractory severe aplastic anaemia. Br J Haematol
31. Kasahara S, Hara T, Itoh H, et al: Hypoplastic myelodysplastic 133:622-627, 2006.
syndromes can be distinguished from acquired aplastic 52. Maciejewski JP, Follmann D, Nakamura R, et al: Increased
anaemia by bone marrow stem cell expression of the tumour frequency of HLA-DR2 in patients with paroxysmal nocturnal
necrosis factor receptor. Br J Haematol 118:181-188, 2002. hemoglobinuria and the PNH/aplastic anemia syndrome.
32. Young NS, Calado RT, Scheinberg P: Current concepts in the Blood 98:3513-3519, 2001.
pathophysiology and treatment of aplastic anemia. Blood 53. Socie G, Mary J-Y, Schrezenmeier H, et al: Granulocyte-
108:2509-2519, 2006. stimulating factor and severe aplastic anemia: a survey by the
33. Solomou EE, Rezvani K, Mielke S, et al: Deficient CD4+ European group for blood and marrow transplant (EBMT).
CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia. Blood 109:2794-2796, 2007.
Blood 110:1603-1606, 2007. 54. Gurion R, Gafter-Gvili A, Paul M, et al: Hematopoietic growth
34. Gidvani V, Ramkissoon S, Sloand EM, et al: Cytokine gene factors in aplastic anemia patients treated with immuno
polymorphisms in acquired bone marrow failure. Am J suppressive therapysystematic review and meta-analysis.
Hematol 82:721-724, 2007. Hematologica 94:712-719, 2009.
35. Hirano N, Butler MO, von Bergwelt-Baildon MS, et al: Auto- 55. Fanconi G: Familiaere infantile perniziosaartige Anaemie
antibodies frequently detected in patients with aplastic (pernizioeses Blutbild und Konstitution). Jahrbuch Kinderheil
anemia. Blood 102:4567-4575, 2003. 117:257-280, 1927.
36. Feng X, Chuhjo T, Sugimori C, et al: Diazepam-binding 56. Tischkowitz MD, Hodgson SV: Fanconi anaemia. J Med Genet
inhibitor-related protein 1: a candidate autoantigen in 40:1-10, 2003.
acquired aplastic anemia patients harboring a minor popula- 57. Green AM, Kupfer GM: Fanconi anemia. Hematol Oncol Clin
tion of paroxysmal nocturnal hemoglobinuria-type cells. North Am 23:193-214, 2009.
Blood 104:2425-2431, 2004. 58. Freedman MH: Inherited forms of bone marrow failure. In
37. Takamatsu H, Feng X, Chuhjo T, et al: Specific antibodies to Hoffman R, Benz EJ, Shattil SJ, et al, editors: Hematology: basic
moesin, a membrane-cytoskeleton linker protein, are fre- principles and practice, ed 5, Philadelphia, 2009, Churchill
quently detected in patients with acquired aplastic anemia. Livingstone, pp 319-357.
Blood 109:2514-2520, 2007. 59. Alter BP: Cancer in Fanconi anemia, 1927-2001. Cancer
38. Ball SE, Gibson FM, Rizzo S, et al: Progressive telomere short- 97:425-440, 2003.
ening in aplastic anemia. Blood 91:3582-3592, 1998. 60. Levitus M, Joenje H, de Winter JP: The Fanconi anemia
39. Calado RT, Young NS: Telomere maintenance and human pathway of genomic maintenance. Cell Oncol 28:3-19,
bone marrow failure. Blood 111:4446-4455, 2008. 2006.
40. Xin Z-T, Beauchamp AD, Calado RT, et al: Functional char- 61. Pinto FO, Leblanc T, Chamousset D, et al: Diagnosis of
acterization of natural telomerase mutations found in patients Fanconi anemia patients with bone marrow failure. Haema-
with hematologic disorders. Blood 109:524-532, 2007. tologica 94:487-495, 2009.
41. Yamaguchi H, Calado RT, Ly H, et al: Mutations in TERT, the 62. Kutler DI, Singh B, Satagopan J, et al: A 20-year perspective
gene for telomerase reverse transcriptase, in aplastic anemia. on the International Fanconi Anemia Registry (IFAR). Blood
N Engl J Med 352:1413-1424, 2005. 101:1249-1256, 2003.
42. Calado RT, Graf SA, Wilkerson KL, et al: Mutations in the 63. Rosenberg PS, Greene MH, Alter BP: Cancer incidence in
SBDS gene in acquired aplastic anemia. Blood 110:1141- persons with Fanconi anemia. Blood 101:822-826, 2003.
1146, 2007. 64. Li X, Leteurtre F, Rocha V, et al: Abnormal telomere metabo-
43. Brummendorf TH, Maciejewski JP, Mak J, et al: Telomere lism in Fanconis anaemia correlates with genomic instability
length in leukocyte subpopulations of patients with aplastic and the probability of developing severe aplastic anaemia. Br
anemia. Blood 97:895-900, 2001. J Haematol 120:836-845, 2003.
44. Camitta BM, Thomas ED, Nathan DG, et al: Severe aplastic 65. Alter BP, Giri N, Savage SA, et al: Cancer in dyskeratosis con-
anemia: a prospective study of the effect of early marrow genita. Blood 113:6549-6557, 2009.
transplantation on acute mortality. Blood 48:63-70, 1976. 66. Savage SA, Alter BP: Dyskeratosis congenita. Hematol Oncol
45. Bacigalupo A, Hows J, Gluckman E, et al: Bone marrow trans- Clin North Am 23:215-231, 2009.
plantation (BMT) versus immunosuppression for the treat- 67. Alter BP, Baerlocher GM, Savage SA, et al: Very short telomere
ment of severe aplastic anaemia (SAA): a report of the EMBT length by flow fluorescence in situ hybridization identifies
SAA working party. Br J Haematol 70:177-182, 1988. patients with dyskeratosis congenita. Blood 110:1439-1447,
46. Sugimori C, Chuhjo T, Feng X, et al: Minor population of 2007.
CD55CD59 blood cells predicts response to immunosup- 68. Goobie S, Popovic M, Morrison J, et al: Shwachman-Diamond
pressive therapy and prognosis in patients with aplastic syndrome with exocrine pancreatic dysfunction and bone
anemia. Blood 107:1308-1314, 2006. marrow failure maps to the centromeric region of chromo-
47. Socie G, Rosenfeld S, Frickhofen N, et al: Late clonal diseases some 7. Am J Hum Genet 68:1048-1054, 2001.
of treated aplastic anemia. Semin Hematol 37:91-101, 2000. 69. Burroughs L, Woolfrey A, Shimamura A: Shwachman-
48. Maciejewski JP, Risitano A, Sloand E, et al: Distinct clinical Diamond syndrome: a review of the clinical presentation,
outcomes for cytogenetic abnormalities evolving from aplas- molecular pathogenesis, diagnosis, and treatment. Hematol
tic anemia. Blood 99:3129-3135, 2002. Oncol Clin North Am 23:233-248, 2009.
49. Kearns WG, Sutton JF, Maciejewski JP, et al: Genomic instabil- 70. Ganapathi KA, Austin KM, Lee C-S, et al: The human
ity in bone marrow failure syndromes. Am J Hematol 76:220- Shwachman-Diamond syndrome protein, SBDS, associates
224, 2004. with ribosomal RNA. Blood 110:1458-1465, 2007.
50. Locasciulli A, Oneto R, Bacigalupo A, et al: Outcome of 71. Austin KM, Gupta ML Jr, Coats SA, et al: Mitotic spindle
patients with acquired aplastic anemia given first line bone destabilization and genomic instability in Shwachman-
marrow transplantation or immunosuppressive treatment in Diamond syndrome. J Clin Invest 118:1511-1518, 2008.
the last decade: a report from the European group for blood 72. Dror Y, Freedman MH: Shwachman-Diamond syndrome.
and marrow transplantation. Haematologica 92:11-18, 2007. Br J Haematol 118:701-713, 2002.
298 PART IV Hematopathology: Erythrocyte Disorders

73. Donadieu J, Leblanc T, Meunier BB, et al: Analysis of risk dyserythropoietic anemia type II. Nat Genet 41(8):936-940,
factors for myelodysplasias, leukemias and death from infec- 2009. Epub June 28, 2009.
tion among patients with congenital neutropenia. Experience 84. Kirchhausen T: Making COPII coats. Cell 129:1325-1336,
of the French Severe Chronic Neutropenia Study Group. 2007.
Hematologica 90:45-53, 2005. 85. Prchal JT: Anemia associated with marrow infiltration. In
74. Maciejewski JP, Tiu RV. Acquired disorders of red cell and Lichtman MA, Beutler E, Kipps TJ, et al, editors: Williams
white cell production. In Hoffman R, Benz EJ, Shattil SJ, hematology, ed 7, New York, 2006, McGraw-Hill, pp 561-563.
et al, editors: Hematology: basic principles and practice, ed 5, 86. Makoni SN, Laber DA: Clinical spectrum of myelophthisis in
Philadelphia, 2009, Churchill Livingstone, pp 395-400. cancer patients. Am J Hematol 76:92-93, 2004.
75. Sawada K, Hirokawa M, Fujishima N: Diagnosis and manage- 87. National Kidney Foundation: KDOQI clinical practice guide-
ment of acquired pure red cell aplasia. Hematol Oncol Clin lines and clinical practice recommendations for anemia in
North Am 23:249-259, 2009. chronic kidney disease. Am J Kidney Dis 47(suppl 3):S1-146,
76. Shaw J, Meeder R: Transient erythroblastopenia of childhood 2006.
in siblings: case report and review of the literature. J Pediatr 88. Marks PW, Rosovsky R: Hematologic manifestations of
Hematol Oncol 29:659-660, 2007. systemic disease: liver and renal disease. In Hoffman R,
77. Lipton JM, Ellis SR: Diamond-Blackfan anemia: diagnosis, Benz EJ, Shattil SJ, et al, editors: Hematology: basic principles
treatment, and molecular pathogenesis. Hematol Oncol Clin and practice, ed 5, Philadelphia, 2009, Churchill Livingstone,
North Am 23:261-282, 2009. pp 2303-2308.
78. Lipton JM, Atsidaftos E, Zyskind I, et al: Improving clinical 89. Coresh J, Selvin E, Stevens LA, et al: Prevalence of chronic
care and elucidating the pathophysiology of Diamond Black- kidney disease in the United States. JAMA 298:2038-2047,
fan anemia: an update from the Diamond Blackfan anemia 2007.
registry. Pediatr Blood Cancer 46:558-564, 2006. 90. Macdougall IC: Role of uremic toxins in exacerbating anemia
79. Renella R, Wood WG: The congenital dyserythropoietic in renal failure. Kidney Int 59(suppl 78):S67-S72, 2001.
anemias. Hematol Oncol Clin North Am 23:283-306, 2009. 91. Caro J: Anemia of chronic renal failure. In Lichtman MA,
80. Heimpel H: Congenital dyserythropoietic anemias: epidemi- Beutler E, Kipps TJ, et al, editors: Williams hematology, ed 7,
ology, clinical significance, and progress in understanding New York, 2006, McGraw-Hill, pp. 449-457.
their pathogenesis. Ann Hematol 83:613-621, 2004. 92. National Kidney Foundation: KDOQI clinical practice guide-
81. Dgany O, Avidan N, Delaunay J, et al: Congenital dyseryth- line and clinical practice recommendations for anemia in
ropoietic anemia type I is caused by mutations in codanin-1. chronic kidney disease: 2007 update of hemoglobin target.
Am J Hum Genet 71:1467-1474, 2002. Am J Kidney Dis 50:479-512, 2007.
82. Noy-Lotan S, Dgany O, Lahmi R, et al: Codanin-1, the protein 93. Wish JB: Assessing iron status: beyond serum ferritin and
encoded by the gene mutated in congenital dyserythropoietic transferrin saturation. Clin J Am Soc Nephrol 1:S4-S8, 2006.
anemia type I (CDAN1), is cell cycle-regulated. Haematologica 94. Bovy C, Gothot A, Delanaye P, et al: Mature erythrocyte
94:629-637, 2009. parameters as new markers of functional iron deficiency in
83. Schwarz K, Iolascon A, Verissimo F, et al: Mutations affecting haemodialysis: sensitivity and specificity. Nephrol Dial Trans-
secretory COPII coat component SEC23B cause congenital plant 22:1156-1162, 2007.
Introduction to Increased
Destruction of Erythrocytes
22
Kathryn Doig

OUTLINE OBJECTIVES
Classification After completion of this chapter, the reader will be able to:
Hemolysis
Normal Bilirubin 1. Define hemolysis and recognize its hallmark clinical 7. Identify laboratory tests that indicate accelerated
Metabolism findings. RBC destruction, explain the physiologic mecha
Normal Plasma 2. Differentiate a hemolytic disorder from hemolytic nisms involved, and interpret the results of each
Hemoglobin Salvage anemia by definition and recognition of laboratory test.
During Intravascular findings. 8. Identify, explain, and interpret the results of labora
Hemolysis 3. Discuss methods of classifying hemolytic anemias tory tests that indicate increased erythropoiesis.
Excessive Extravascular and apply the classification to an unfamiliar anemia. 9. Differentiate between hemolytic anemias and other
Hemolysis 4. Describe the processes of intravascular (fragmen causes of increased erythropoiesis given laboratory
Excessive Intravascular tation) and extravascular (macrophage-mediated) or clinical information.
Hemolysis
hemolysis, emphasizing sites of hemolysis for each 10. Describe the mechanisms that salvage hemoglobin
Clinical Features
and associating the clinical findings of each. during intravascular hemolysis.
Laboratory Findings
Tests of Accelerated 5. Differentiate intravascular and extravascular hemo 11. Explain the rationale for tests of the hemoglobin
Red Blood Cell lysis when given clinical and laboratory findings. salvage mechanisms and interpret the results of
Destruction 6. Describe the process of normal red blood cell (RBC)/ each.
Tests of Increased hemoglobin catabolism, emphasizing the process for
Erythropoiesis catabolism of protoporphyrin.
Laboratory Tests
to Determine
Specific Hemolytic CASE STUDY
Processes After studying the material in this chapter, the reader should be able to respond to the following
Differential Diagnosis case study:

A 34-year-old woman was admitted to the hospital for a vaginal hysterectomy. Except for excessive
menstrual bleeding, she was in otherwise good health, and all of her preoperative laboratory test
results were within the reference range. There was no excessive blood loss during or after surgery,
and recovery was uneventful except for some cramping, for which the patient received ibuprofen.
Three days after surgery, a CBC revealed a hemoglobin level of 5.8 g/dL. Subsequently, the
patient began to experience abdominal pain and passed root beercolored urine.
1. What process is indicated by the root beercolored urine?
2. What laboratory tests can be used to differentiate the cause of the hemolysis?
3. Based on the patients clinical presentation, predict the results expected for each test listed for
question 2.

T
his chapter presents an overview of the hemolytic reduced number of cells results in reduced tissue oxygenation
process and provides a foundation that is applicable in and increased erythropoietin production by the kidney. When
the following chapters on red blood cell (RBC) disor- the patient is otherwise healthy, the bone marrow responds by
ders. The term hemolysis or hemolytic disorder refers to increased accelerating erythrocyte production, which leads to reticulo
destruction (i.e., lysis) of RBCs, shortening their life span. The cytosis. A hemolytic process is present without anemia if the

299
300 PART IV Hematopathology: Erythrocyte Disorders

bone marrow is able to compensate by accelerating RBC pro- a noncomprehensive list of hemolytic anemias. This chapter
duction sufficiently to replace the RBCs lost through hemoly- focuses on the distinction between intravascular and extravas-
sis. Healthy bone marrow can increase its production of RBCs cular hemolytic conditions. The other classifying schemes are
by six to eight times normal1; therefore, significant RBC destruc- summarized here briefly and are used to organize the chapters
tion must occur before an anemia develops. A hemolytic that follow.
anemia results when the rate of RBC destruction exceeds the Acute versus chronic hemolysis delineates the clinical presenta-
increased rate of RBC production. tion. Acute hemolysis has a rapid onset and is isolated (sudden),
episodic, or paroxysmic, as in paroxysmal cold hemoglobinuria or
paroxysmal nocturnal hemoglobinuria. Patients with paroxysmal
CLASSIFICATION
cold hemoglobinuria experience hemolysis only after exposure
Many anemias have a hemolytic component, including the to cold, and patients with paroxysmal nocturnal hemoglobin-
anemia associated with vitamin B12 or folate deficiency and the uria experience hemolysis while sleeping. Whatever the cause,
anemia of chronic inflammation, renal disease, and iron defi- acute hemolysis either disappears or subsides between epi-
ciency. In these conditions, the hemolysis alone does not cause sodes, during which time the patients condition returns to
anemia, and so they are not considered hemolytic disorders. normal. A hemolytic transfusion reaction is an example of a
Rather, these anemias develop as a result of the inability of the single acute incident.
bone marrow to increase production of RBCs. Because hemo- Chronic hemolysis may not be evident if the bone marrow
lysis is not the primary underlying cause, these disorders are is able to compensate, but may be punctuated over time with
considered anemias with a secondary hemolytic component. hemolytic crises that cause anemia. Glucose-6-phosphate dehy-
When hemolysis is the primary feature, anemias can be drogenase deficiency is such a condition. RBC life span is chroni
classified as follows: cally shortened, but bone marrow compensation prevents
Acute versus chronic anemia. When the cells are challenged with oxidizing agents
Inherited versus acquired such as antimalarial drugs, a dramatic acute hemolytic event
Intrinsic versus extrinsic occurs. When the drug is withdrawn, compensation returns.
Intravascular (fragmentation) versus extravascular Other chronic conditions result in anemia that is so severe
(macrophage-mediated) that the bone marrow cannot generate cells fast enough to
Every hemolytic condition can be classified according to compensate for the anemia. Thalassemia is an example of such
each of these descriptors. Table 22-1 shows this and provides a condition. Although cell production is brisk, each cell

TABLE 22-1 Classification of Selected Hemolytic Anemias by Primary Cause and Type of Hemolysis

Predominantly Intravascular Hemolysis Predominantly Extravascular Hemolysis


Agents from Outside the RBC
Immune hemolysis: cold antibody Immune hemolysis: warm antibody
Microangiopathic hemolysis Drugs
Infectious agents, as in malaria
Extrinsic defects

Acquired conditions
Thermal injury
Chemicals/drugs
Venoms
Prosthetic heart valves

Membrane Abnormalities
Spur cell anemia of severe liver disease
Paroxysmal nocturnal hemoglobinuria
Hereditary membrane defects such as
Hereditary conditions
Intrinsic defects

spherocytosis

Abnormalities of the RBC Interior


Enzyme defects such as G6PD deficiency
Globin abnormalities such as sickle cell, thalassemia

Green text indicates acute or episodic hemolysis.


Red text indicates chronic hemolysis.
Some conditions may exhibit mixed presentations under certain circumstances. It is evident that most hereditary conditions lead to chronic hemolysis, whereas acquired conditions
are more often acute. Furthermore, the intrinsic red cell defects typically are due to hereditary conditions, whereas extrinsic factors typically lead to acquired hemolytic
disorders.
G6PD, Glucose-6-phosphate dehydrogenase; RBC, red blood cell.
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 301

possesses an inadequate complement of one type of globin, and thus are outside the bloodstream. This classification is
and hemoglobin (Hb) production is decreased. As a result the useful because laboratory testing readily distinguishes the two
oxygen-carrying capacity of the blood is chronically low. Cells types. However, the specific cause of the hemolysis must still
lyse in thalassemia because excess normal globin chains pre- be determined for appropriate treatment.
cipitate inside the cells, which leads to hemolysis and chronic
anemia.
HEMOLYSIS
Inherited hemolytic conditions, such as thalassemia, are
passed to offspring by mutant genes from the parents. Acquired Normal Bilirubin Metabolism
hemolytic disorders develop in individuals who were previ- Detection of hemolysis depends partly on detection of RBC
ously hematologically normal but acquire an agent or condi- breakdown products. A prominent product is bilirubin. The
tion that lyses RBCs. Infectious diseases such as malaria are an process of normal bilirubin production is described to clarify
example. the relationship between hemolysis and increased bilirubin
The hemolytic conditions are also classified as involving levels.
intrinsic or extrinsic RBC defects, with the latter caused by the The story of bilirubin production is, in part, a story of iron
action of external agents. This is the classification scheme used salvage. The body salvages and recycles iron like a precious
for subsequent chapters in this book. Examples of intrinsic metal. If this were not true, there would perhaps be a hemo-
hemolytic disorders are abnormalities of RBC membranes, globin and iron excretion system, but there is not. Because iron
enzymatic pathways, or the hemoglobin molecule. With intrin- must be salvaged, however, there is a process for saving heme
sic defects, if the RBCs of the affected patient were to be trans- and globin. Bilirubin is the excretory product for the protopor-
fused into a healthy individual, they would still have a phyrin component of heme.
shortened life span, because the defect is in the RBC. If normal RBCs live approximately 120 days. During this time, they
RBCs are transfused into a patient who has an intrinsic defect, undergo various metabolic and chemical changes, which result
the transfused cells have a normal life span, because the trans- in a loss of deformability. Under normal circumstances, mac-
fused cells are normal. rophages of the mononuclear phagocyte system (or reticuloen-
Extrinsic hemolytic conditions are those that arise from dothelial system) recognize these changes and phagocytize the
outside the RBC, typically substances in the plasma or condi- aged erythrocytes (see Chapter 8). The organs of this system
tions affecting the anatomy of the circulatory system. Even include the spleen, bone marrow, liver, lymph nodes, and
though malaria protozoa and other infectious agents are within circulating monocytes, but it is primarily the spleen and liver
the RBC, they are classified as extrinsic because the RBC was that process senescent RBCs. The macrophages in the spleen
normal until it was invaded by an outside agent. An antibody (littoral cells) are especially sensitive to subtle RBC changes,
against RBC antigens and a prosthetic heart valve are examples whereas the macrophages of the liver (Kupffer cells) detect and
of noninfectious extrinsic agents. In extrinsic hemolysis, cross- destroy more severely damaged RBCs. This is an extravascular
transfusion studies have shown that the patients RBCs have a hemolytic process. Hemolysis occurs within macrophages; thus
normal life span in the bloodstream of a normal individual, macrophage-mediated hemolysis is a more precise term.
but normal cells are lysed more rapidly in the patients circula- The majority of RBC degradation occurs extravascularly as
tion. These studies confirm that something outside the RBCs enzymes of the macrophage lyse the phagocytized erythrocytes
is causing the hemolysis. (Of course, in the case of intracellular (Figure 22-1). Hemoglobin is hydrolyzed into heme and
parasites, the cross-transfusion study is not applicable.) Most globin; the latter is further degraded into amino acids that
intrinsic defects are inherited; most extrinsic ones are acquired return to the amino acid pool. Iron is released from the heme,
(see Table 22-1). A few exceptions exist, such as paroxysmal returned to the plasma via ferroportin, bound to its protein
nocturnal hemoglobinuria, an acquired disorder involving an carrier molecule (transferrin), and recycled to developing
intrinsic defect (see Chapter 23). RBCs. The remaining protoporphyrin is degraded through a
Intrinsic disorders are subclassified as membrane defects, series of biochemical reactions in different tissues and organs.
enzyme defects, and hemoglobinopathies. Extrinsic hemolysis Figure 22-1 illustrates protoporphyrin catabolism. While
may be immunohemolytic, traumatic, or microangiopathic, or protoporphyrin is inside the macrophage, heme oxygenase acts
may be caused by infectious agents, chemical agents (drugs and on it, breaking the porphyrin ring to yield a linear molecule,
venoms), or physical agents (see Table 22-1). biliverdin. The lungs excrete a by-product of that reaction,
Another classification scheme is based on the site of hemo- carbon monoxide. The green biliverdin is reduced to bilirubin,
lysis and the general mechanism of lysis. Intravascular hemoly- a nonpolar yellow molecule that is secreted into the plasma
sis occurs by fragmentation. Although this takes place most (Box 22-1). Because it is hydrophobic, this bilirubin must
often within the bloodstream, cells can lyse by fragmentation bind to albumin to be transported in plasma to the liver.
in the spleen and bone marrow as well. Extravascular hemolysis The bilirubin-albumin complex enters the liver parenchymal
occurs when RBCs are engulfed by macrophages and lysed by cells, where the bilirubin dissociates from the albumin and
their digestive enzymes. The designation extravascular, meaning is joined (i.e., conjugated) with two molecules of glucuronic
outside the vessels, can refer either to lysis within the macro- acid by glucuronyl transferase to form bilirubin diglucuro-
phage and not in the bloodstream, or to the fact that most of nide.2 The addition of the two sugar acid molecules makes
the macrophages are in tissues, chiefly the spleen and the liver, the molecule polar and water soluble. Bilirubin diglucuronide
302 PART IV Hematopathology: Erythrocyte Disorders

Hb Hemoglobin Urobilinogen

Red blood cell Unconjugated bilirubin bound to albumin

Conjugated bilirubin

Unconjugated bilirubin formed


Heart
CO
to lung
Macrophage
Hb

Heme + globin Spleen


Heme oxygenase
Biliverdin

Unconjugated
Bilirubin
Bound to
Liver albumin

Kidney
Unconjugated
bilirubin
can't pass Paren-
through chymal
glomerulus cell Macrophage
due to the
albumin

Bone
marrow
Albumin split off
Portal Bile
duct Unconjugated bilirubin
vein +
Urine Glucuronic acid
Glucuronyl
Trace urobilinogen, transferase
no bilirubin
Intestines Conjugated bilirubin

Stool Bacteria converting


Urobilinogen & bilirubin to urobilinogen
urobilin,
no bilirubin

Figure 22-1 Normal catabolism of aged red blood cells.

is also called conjugated bilirubin. The bilirubin originally


released from macrophages that lacks these sugars is termed
BOX 22-1 Visualizing the Color Changes of
unconjugated.
Hemoglobin Degradation
Conjugated bilirubin is excreted as bile acid into the
The degradation of heme can be seen in bruises in fair-skinned
intestines. There it assists with the emulsification of fats for
individuals or in the sclera of the eye after a vascular bleed. The
absorption from the diet. Conjugated bilirubin is oxidized by
same process that macrophages facilitate can occur in tissues. At
gut bacteria into various water-soluble compounds, collectively
first, the extravasated but deoxygenated blood gives the injury the
called urobilinogen. Most urobilinogen is oxidized further
purple-red appearance of hemoglobin. As the hemoglobin is
to stercobilin and similar compounds that give the brown
degraded, the color changes to a greenish hue due to biliverdin, but
color to stool, which is the ultimate route for excretion of
ultimately it becomes yellow due to bilirubin.
protoporphyrin.
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 303
Because they are water soluble, some conjugated bilirubin hemoglobin and iron can cause oxidative damage to cells.
and urobilinogen molecules are absorbed from the intestines Several mechanisms exist to salvage hemoglobin iron and
and can be detected in the plasma. The portal circulation, the prevent oxidation reactions, and are collectively called the
blood vessels that surround the intestines to absorb nutrients, haptoglobin-hemopexin-methemalbumin system (Figure 22-2).
collect these bile products by osmosis. The portal circulation When free in the plasma, hemoglobin exists as / dimers
carries blood directly to the liver, so most of the absorbed bound to a liver-produced plasma protein called haptoglobin.
conjugated bilirubin and urobilinogen is recycled directly into This is the first mechanism of hemoglobin iron salvage. By
the bile again. Some remains in the plasma, however, and is binding to haptoglobin, hemoglobin avoids filtration at the
filtered by the renal glomerulus and excreted in the urine. glomerulus and is saved from urinary loss. The complex is
Direct bilirubin is virtually undetectable in urine, but a measur- carried to the liver, where the haptoglobin-hemoglobin
able amount of urobilinogen can be expected normally. The complex is bound to macrophage receptors and internalized
yellow color of urine is due to urobilin, a derivative of urobi-
linogen, that is also water soluble and absorbed into the portal
circulation. Urobilinogen is colorless.
BOX 22-2 Laboratory Impact of Significant
Hemoglobinemia
Normal Plasma Hemoglobin Salvage During The results of a routine complete blood count are unreliable for
Intravascular Hemolysis patients with significant hemoglobinemia. Under normal circum
Intravascular hemolysis is trauma to the RBC membrane that
stances, the measured hemoglobin represents the hemoglobin
causes a breach sufficient for the cell contents, chiefly hemo-
present inside the red blood cells. For individuals with hemoglobine
globin, to spill directly into plasma (Box 22-2). Approximately
mia, the intracellular hemoglobin and the plasma hemoglobin both
10% to 20% of normal RBC destruction is intravascular,3
are measured. The hemoglobin value therefore is falsely elevated.
secondary to turbulence and anatomic restrictions in the
An unrealistically high value for mean cell hemoglobin concentration
vasculature.
may provide a clue to this problem, which can be remedied in several
Because hemoglobin is filtered by the kidney, iron could
ways (see Chapters 14 and 39).
be lost in normal intravascular hemolysis. In addition, free

Lysed RBC
in blood vessel Globin
Free Blood
Methemoglobin vessel
hemoglobin

Albumin
Metheme Methemalbumin
Haptoglobin Hemopexin
/ dimers Hemoglobin/haptoglobin

Metheme/hemopexin
Liver
Transferrin
Globin Amino acids
CO to lungs
Iron Heme
Kidney Renal tubular Heme oxygenase
epithelial Biliverdin
cell with
hemosiderin
Bilirubin + glucuronic acid
Glucuronyl
transferase
Bilirubin diglucuronide

Urine Intestines
Hemoglobinuria
Hemosiderinuria Urobilinogen Bacteria Fecal
Increased urobilinogen urobilinogen

Figure 22-2 Iron salvage mechanisms during intravascular hemolysis. CO, Carbon monoxide; RBC, red blood cell.
304 PART IV Hematopathology: Erythrocyte Disorders

into the macrophage.4 Inside the macrophage, iron is salvaged, hemoglobin formed in various anemias, bind to the inner
and the remaining protoporphyrin is converted to bilirubin, surface of the RBC membrane, producing changes to the exte-
just as though the RBC had been ingested by the macrophage. rior of the membrane that can be detected by macrophages.
If lysis is accelerated, haptoglobin is depleted, because the When something causes increased formation of Heinz bodies
livers production of haptoglobin does not increase in response or other intracellular inclusions, the cells are removed from the
to the increased hemolysis and is typically adequate to salvage circulation prematurely by macrophages. A similar process
only a normal amount of plasma hemoglobin. occurs when intracellular parasites are present or when com-
A secondary mechanism of iron salvage involves hemopexin. plement or immunoglobulins are on the surface of the RBC.
In hemoglobin that is not bound by haptoglobin, the iron When the RBC is ingested, it is lysed within a phagosome,
becomes oxidized, forming methemoglobin. The heme mole- and the contents are processed within the macrophage as
cule (actually metheme) dissociates from the globin and binds described previously. The contents of the RBC are not detected
to another liver-produced plasma protein, hemopexin.5,6 This in plasma because it is lysed inside the macrophage, and the
binding also saves the iron from urinary loss and prevents contents are degraded therehence the designation extravascu-
oxidation of cell membranes. Hemopexin-metheme binds to lar hemolysis. Extravascular hemolysis occurs most often in the
hepatocyte receptors and is internalized. There the iron is sal- spleen and liver, where the macrophages possess receptors for
vaged, and the protoporphyrin is converted to bilirubin, as in the markers that identify defective RBCs, principally immuno-
a macrophage. In contrast to haptoglobin, hemopexin is recy- globulins and complement.
cled to the plasma from the hepatocyte, so a single hemopexin Sometimes the macrophage ingests a portion of the mem-
molecule collects and salvages more free metheme molecules. brane, leaving the remainder to reseal. Little, if any, cytoplas-
Although its production is constant, because it is recycled to mic volume is lost, but with less membrane, the cell becomes
the plasma, levels of hemopexin do not fall dramatically during a spherocyte, the characteristic shape change associated with
periods when there is an increase in intravascular hemolysis. extravascular hemolysis. Although the spherocyte reenters the
A third mechanism of iron salvage is the metheme-albumin circulation, its survival is shortened because of its rigidity and
system. Albumin acts a carrier for many molecules. Metheme inability to traverse the splenic sieve during subsequent pas-
can bind to albumin when hemopexin is saturated, which sages through the red pulp. It may become trapped against the
further reduces urinary loss. This is probably a temporary basement membrane of the splenic sinus and be fully ingested
holding state for the metheme, which transfers to hemopexin by a macrophage, or it may lyse mechanically and in so doing
because it has a higher binding affinity for metheme than does contribute an intravascular (i.e., fragmentation) component to
albumin.7 It is then transported to the liver for processing. what is otherwise an extravascular process.
If the previous systems are overloaded, the excess hemoglo- In extravascular hemolytic anemias, the plasma bilirubin
bin or heme iron will be filtered into the urine. Iron still can level rises as RBCs lyse prematurely. The plasma bilirubin is
be retained in the kidney, however.8,9 Some hemoglobin or largely unconjugated. As long as the liver is healthy, it processes
(met)heme that enters the filtrate is reabsorbed, not by specific the increased load of unconjugated bilirubin, producing con-
carriers but as part of the general filtrate in the proximal tubule. jugated bilirubin that enters the intestine. Increased urobilino-
When inside the renal tubular cell, the iron is separated from gen forms in the intestines and is subsequently absorbed by
the protoporphyrin. There it is stored as ferritin or hemosi the portal circulation and excreted by the kidney. As a result,
derin. The role of the kidney in salvaging iron and returning it increased urobilinogen is detectable in the urine. Although
to the circulation is uncertain at this time. Although carrier there is an increase in unconjugated bilirubin in the plasma,
proteins like divalent metal transporter 1 and ferroportin have none of it appears in the urine because it is bound to albumin
been identified in the kidney of experimental mice, their local- and cannot pass through the glomerulus. These findings are
ization, function, and regulation are still not clear. When the summarized in Table 22-2.
amount of hemoglobin presented to the kidney exceeds what
can be reabsorbed, the excess is then detectable in the urine.
EXCESSIVE INTRAVASCULAR HEMOLYSIS
Although intravascular hemolysis is a minor component of
EXCESSIVE EXTRAVASCULAR HEMOLYSIS
normal RBC destruction, it can be a major feature of pathologic
Many hemolytic anemias are a result of increased extravascular processes (Figure 22-4). Dramatic examples of intravascular
hemolysis (Figure 22-3) during which more than the usual hemolysis are the traumatic, physical lysis of RBCs caused by
number of RBCs are removed from the circulation daily. Under prosthetic heart valves and the exit of mature intracellular RBC
normal circumstances, senescent RBCs display surface markers parasites, such as malaria protozoa, by bursting out of the cell.
that identify them to macrophages as aged cells requiring In these instances, the intravascular destruction of RBCs causes
removal (see Chapter 8). Pathologic processes also lead to profound anemias.
expression of abnormal markers, so that cells are identified and Intravascular hemolysis is fragmentation of the RBCs that
removed. If the number of affected cells increases beyond the releases detectable hemoglobin into the plasma. Characteristic
quantity normally removed each day due to senescence, and if laboratory findings are hemoglobinemia, hemoglobinuria, and
the bone marrow cannot compensate, then anemia develops. hemosiderinuria (see Table 22-2). In addition, methemalbumin
As an example, Heinz bodies, aggregates of denatured and hemopexin-heme are detected in plasma, and the
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 305

Urobilinogen

Red blood cell Unconjugated bilirubin bound to albumin

Conjugated bilirubin

In blood: increased unconjugated


Heart bilirubin and urobilinogen

Increased
destruction of
RBC, causing more Spleen
unconjugated
bilirubin

Liver

Kidney
Unconjugated
bilirubin
can't pass
through
glomerulus
due to the
albumin

Bone
marrow

Portal Bile
vein duct Increased amount of
unconjugated bilirubin,
Urine so increased amount of
Increased bilirubin being conjugated
urobilinogen
Intestines

Stool Increased amount


Increased of urobilinogen formed
urobilinogen

Figure 22-3 Increased extravascular hemolysis. RBC, Red blood cell.

haptoglobin level is decreased. Plasma bilirubin (largely During excess intravascular hemolysis, plasma haptoglobin
unconjugated) and elevated levels of urinary urobilinogen are is rapidly depleted. Levels of hemopexin also decline, but not
also measurable but not immediately, because time is needed as significantly as haptoglobin levels, because of its recycling.
to produce these products. The time course of these findings The hemopexin level is decreased during severe hemolysis,
assists with the differential diagnosis (see later). but it is not often measured. Levels of the hemopexin-heme
In a properly collected specimen, normal intravascular complex, however, increase. Methemalbumin can also be
hemolysis produces a plasma hemoglobin level of less than detected in the plasma, although it too is seldom measured.
1mg/dL.10 Plasma does not become visibly red until the The presence of methemalbumin and hemopexin-heme, which
plasma hemoglobin level is at 50mg/dL.10 Typical values impart a coffee-brown color to plasma, strongly suggests intra-
during hemolytic processes can range from 15 to 100mg/dL, vascular hemolysis.12,13 Classically, these pigments have been
so an increase in plasma hemoglobin may not always be detected directly in plasma spectrophotometrically at 620 to
visible.11 630nm and do not disappear with the addition of hydrogen
306 PART IV Hematopathology: Erythrocyte Disorders

TABLE 22-2 Comparison of Laboratory Findings Indicating Accelerated Red Blood Cell (RBC) Destruction in Intravascular
versus Extravascular Hemolysis
Test Sample Result Intravascular Extravascular
Serum Increased total bilirubin X X
Increased indirect (unconjugated) bilirubin X X
Normal direct (conjugated) bilirubin X X
Increased lactate dehydrogenase activity (RBC fraction) X x
Decreased haptoglobin X x
Increased free hemoglobin X x
Decreased hemopexin X x
Urine Increased urobilinogen X X
Presence of free hemoglobin X
Presence of methemoglobin X
Positive reaction with Prussian blue staining of urine sediment X
Anticoagulated whole blood Decreased hemoglobin, hematocrit, RBCs X X
Presence of schistocytes X
Presence of spherocytes X
Decreased glycated hemoglobin X X
Special tests Increased endogenous carbon monoxide X X
Decreased erythrocyte life span X X

X, Typically expected; x, minor component.

peroxide, as does methemoglobin. If ammonium sulfide is the skin of fair-skinned individuals. Jaundice refers to the color
added (Schumm test), the 620- to 630-nm band disappears, of the skin and sclera, whereas icterus describes plasma. An
and a band at 558nm forms.14 More recently, enzyme-linked increase in plasma bilirubin and subsequent jaundice can
immunosorbent assays have been developed for hemopexin. occur in other conditions besides hemolysis, but if the jaundice
In addition, if the plasma hemoglobin is sufficiently ele- is the result of hemolysis, it is called hemolytic jaundice. It also
vated, hemoglobin is detectable in the urine (hemoglobin- may be called prehepatic jaundice, which reflects the predomi-
uria). For example, when an acute intravascular hemolytic nance of unconjugated bilirubin.
transfusion reaction is suspected, a urine sample is tested The frequency or constancy of jaundice provides clues to
promptly for hemoglobin using the urinalysis reagent strip. the cause. In glucose-6-phosphate dehydrogenase deficiency,
Hemoglobin may give a dark red or brown color (due to met- for example, hemolysis is periodic, appearing during a crisis.
hemoglobin) to the urine if the amount is sufficiently high, In thalassemia, jaundice is chronic. Jaundice may not be
and thus the urine may be described as looking like root beer evident if hemolysis is minimal and the liver is able to process
or beer. In chronic intravascular hemolysis, iron absorbed into the additional bilirubin, as is often the case in hereditary
renal tubular epithelial cells may be visualized. These cells elliptocytosis.
slough into the urine and appear in the sediment, and they can Some signs differentiate chronic from acute hemolysis.
be stained with Prussian blue. This is an easy and inexpensive Splenomegaly can develop, particularly with chronic extravas-
way to detect intravascular hemolysis.11 These findings are cular hemolytic processes. Gallstones (cholelithiasis) can occur
summarized in Table 22-2. whenever hemolysis is chronic; the constantly increased
amount of bilirubin in the bile leads to the formation of the
stones.15 When hemolysis is chronic in children, the persistent
CLINICAL FEATURES
compensatory bone marrow hyperplasia can lead to bone
The clinical findings typical of hemolysis may be prominent if deformities, because the bones are still forming (see Figure
the hemolytic process is the primary cause of anemia. For 27-3). For patients in whom an acquired, acute hemolytic
patients in whom hemolysis is secondary, however, other process develops, the associated malaise, aches, vomiting, and
clinical features may be more noticeable. If hemolysis is suf- possible fever may cause it to be confused with an acute infec-
ficient to result in anemia, patients experience the general tious process. Profound prostration and shock may develop,
symptoms of fatigue, dyspnea, and dizziness to a degree that particularly with acute intravascular hemolysis. Flank pain, oli-
is consistent with the severity and rate of development of the guria, or anuria develops, which leads to acute renal failure.
anemia. The associated signs of pallor and tachycardia can Other clinical features may offer a clue as to whether hemo-
be expected. lysis is intravascular or extravascular. In particular, brown
Increase in bilirubin gives a yellow tinge to the plasma and urine, associated with (met)hemoglobinuria, points to an
body tissues. It is readily evident in the sclera of the eyes and intravascular hemolytic process.
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 307

Red blood cell (RBC) lysing Urobilinogen

Hemoglobin Unconjugated bilirubin bound to albumin

Conjugated bilirubin

Blood
Increased unconjugated bilirubin
Heart Increased urobilinogen

Macrophage
Hemoglobin
breaking down

Unconjugated Spleen
Lysis of RBC bilirubin
in blood vessel formed
to form free
hemoglobin Liver
parenchymal
cell RBC
Liver lysis in
vessel
Kidney
Unconjugated
bilirubin
can't pass
through
glomerulus
Macrophage
due to the
albumin

Bone
marrow
Renal tubular epithelial Albumin split off
cell with hemosiderin Portal Bile Conjugated with glucuronide
vein duct (conjugated bilirubin)
Urine
Increased urobilinogen
Hemoglobinuria
Hemosiderinuria Intestines

Stool Bacteria converting


Urobilinogen & bilirubin to urobilinogen
urobilin,
no bilirubin

Figure 22-4 Increased intravascular hemolysis.

expected to be seen with extravascular hemolysis (see Table


LABORATORY FINDINGS
22-2), whereas fragmented cells, or schistocytes, are noted with
In patients with the clinical features of hemolytic anemia, labo- intravascular hemolysis. Other clues to the particular cause of
ratory tests typically show evidence of increased erythrocyte the hemolysis may be present in the morphology, such as ring
destruction and the compensatory increase in the rate of eryth- forms in malaria, sickle cells in sickle cell anemia, target cells
ropoiesis. Other tests that are specific to a particular diagnosis and microcytes in thalassemia, and spherocytes in hereditary
also may be indicated. spherocytosis or immune-related hemolysis.
In either intravascular or extravascular hemolysis, the
Tests of Accelerated Red Blood increased rate of hemoglobin catabolism results in increased
Cell Destruction amounts of the principal by-products, bilirubin and carbon
For a patient with a hemolytic process, a complete blood count monoxide. Most of the bilirubin in bilirubinemia is unconju-
(CBC) may provide clues to the cause. Spherocytes can be gated (also called indirect bilirubin) (Box 22-3). If liver function
308 PART IV Hematopathology: Erythrocyte Disorders

results, however. Low values suggest hemolysis but may be due


BOX 22-3 Laboratory Testing for Serum Bilirubin instead to impaired synthesis caused by liver disease. Alterna-
Because bilirubin occurs in two forms, conjugated and unconjugated, tively, a patient with hemolysis may have a relatively normal
the total bilirubin level in serum is the total of the two forms: total haptoglobin level if there is also a complicating infection or
serum bilirubin = unconjugated serum bilirubin + conjugated serum inflammation, because haptoglobin is an acute phase reactant.
bilirubin (each expressed in mg/dL). Tests to determine the rate of endogenous carbon monoxide
Normally, there is relatively little total bilirubin, and most of it is production have been developed, because carbon monoxide is
composed of the unconjugated form in transit from the macro produced in the first step of heme breakdown. Values of 2 to
phages, where it was produced, to the liver. The small amount of 10 times the normal rate have been detected in some patients
direct bilirubin in the plasma has been absorbed from the intestine with hemolytic anemia, but the methods are too complex for
by the portal circulation. most clinical laboratories.17
Typical reference ranges are as follows: Other laboratory test results are incidentally abnormal.
Total serum bilirubin level = 0.5-1.0mg/dL Serum lactate dehydrogenase activity is often increased in
Direct (conjugated) serum bilirubin level = 0-0.2mg/dL patients with intravascular hemolysis, but other conditions,
Indirect (unconjugated) serum bilirubin level = 0-0.8mg/dL such as myocardial infarction or liver disease, also can cause
Conjugated bilirubin is a polar molecule and reacts well in the increases.18 Although enzyme fractionation points to an RBC
water-based spectrophotometic assay that uses diazotized sulfanilic origin of the increase, this test is generally not needed. Rather,
acid. Unconjugated bilirubin does not react well in this system unless when other results point to hemolysis, one should expect an
alcohol is added to promote its solubility in water. Conjugated bili increase in the levels of lactate dehydrogenase and other RBC
rubin also is called direct bilirubin because it reacts directly with the enzymes and should not be misled into assuming that there is
reagent, and unconjugated bilirubin is called indirect because it has liver damage (see Table 22-2).
to be solubilized first.* When alcohol is added to the test system, General evidence of reduced RBC survival can be gleaned
however, both the direct and indirect forms react. In practice, the by measuring glycated hemoglobin (by the Hb A1c test).19 Gly-
total bilirubin level is measured by adding a solubilizing reagent. In cated hemoglobin increases over the life of a cell as it is exposed
a separate test, the direct bilirubin is measured alone without addi to plasma glucose. Glycated hemoglobin level is usually
tion of the solubilizing agent. The indirect bilirubin level is calculated decreased in hemolytic disease, because the cells have less
by subtracting the direct value from the total value: total serum bili exposure to plasma glucose before lysis. The mean reference
rubin direct serum bilirubin = indirect serum bilirubin. value for glycated hemoglobin is 6.7%; in a hemolytic process,
*Van den Bergh AA, Muller P: ber eine direkte und eine indirekte diazoreaktion the mean value is 3.9%.19 The magnitude of the decrease is
aus bilirubin, Biochem Z 77:90, 1916; and Hutchinson DW, Johnson B, Knell AJ: related to the magnitude of the hemolytic process over the
The reaction between bilirubin and aromatic diazo compounds, Biochem J previous 4- to 8-week period. Glycated hemoglobin level is not
127:907-908, 1972. a reliable indicator of shortened RBC survival in patients with
diabetes mellitus because of the increased rate of glycation with
is normal, conjugated bilirubin is formed and excreted as uro- elevated blood glucose levels.
bilinogen in the stool, and the serum level of conjugated bili- A more exact RBC survival assay uses random labeling of
rubin (also called direct bilirubin) remains within the reference blood with chromium radioisotope. This is the reference
range. No bilirubin is detected in the urine, because unconju- method for RBC survival studies published by the International
gated bilirubin is bound to albumin and the complex is not Committee for Standardization in Haematology.20 A sample of
filtered by the glomerulus. Urinary urobilinogen level may be blood is collected, mixed with the isotope, and returned to the
increased, however, because there is increased urobilinogen in patient. The labeled cells are of all ages, reflecting normal
the stool, and more than usual amounts are absorbed by the peripheral blood. This method differs from cohort labeling in
portal circulation. Serum bilirubin values can be misleadingly which RBCs are labeled with radioactive iron as they are pro-
low, because the amount of bilirubin in the blood depends on duced in the bone marrow, so that the labeled cells are generally
the rate of RBC catabolism and hepatic function. In some of the same age. In both methods, the disappearance of the label
patients with hemolytic anemia, if the rate of hemolysis is low from the blood is measured over time. As measured using the
and liver function is normal, the total serum bilirubin level is random chromium labeling technique, the normal half-time
within the reference range. Quantitative measurements of fecal of chromium is 25 to 32 days.20 A half-time of 20 to 25 days
urobilinogen would demonstrate an increase, however. suggests mild hemolysis; 15 to 20 days, moderate hemolysis;
A substantial decline in serum haptoglobin level indicates and less than 15 days, severe hemolysis. Erythrocyte life span
intravascular hemolysis. In what is mostly an extravascular determinations are time consuming and expensive, require the
hemolysis, there can be a minor component of intravascular use of radioactive isotopes, and are rarely necessary for diagno-
lysis involving spherical cells that are fragile, so a modest sis except in patients with difficult diagnostic problems.
decline in haptoglobin level can be seen. Suffice it to say that,
whenever the level of hemoglobin in the plasma increases, Tests of Increased Erythropoiesis
haptoglobin level declines. In one study, a low haptoglobin If the bone marrow is healthy, the hypoxia associated with
level indicated an 87% probability of hemolytic disease.16 hemolysis leads to increased erythropoiesis. Recognition of
Haptoglobin tests are prone to false positive and false negative this increase may be a first clue to the presence of a hemolytic
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 309

TABLE 22-3 Hematologic Findings Indicating TABLE 22-4 Morphologic Abnormalities Associated
Accelerated Red Blood Cell (RBC) Production with Hemolytic Anemia
Specimen Findings RBC Morphology Hemolytic Disorders
Anticoagulated Increased reticulocyte count Spherocytes Hereditary spherocytosis, warm hemolytic
peripheral blood Rising mean cell volume (compared with baseline) anemia, thermal injury to RBCs
Polychromasia, nucleated RBCs Elliptocytes (ovalocytes) Hereditary elliptocytosis
Bone marrow Erythroid hyperplasia Acanthocytes Abetalipoproteinemia, severe liver disease
Burr cells Pyruvate kinase deficiency, uremia
Schistocytes Microangiopathic hemolytic anemia
process. Laboratory findings indicating increased erythropoie- Erythrophagocytosis Damage to RBC surface, especially due to
sis are listed in Table 22-3. These findings are persistently complement-fixing antibodies
present in chronic hemolytic disease and are evident within 3 RBC agglutination Cold agglutinins, immunohemolytic disease
to 6 days after an acute hemolytic episode. Increased erythro-
poiesis is not unique to hemolytic anemias and is not diagnos- RBC, Red blood cell.
tic. Similar results are expected after hemorrhage and with
successful specific therapy for anemia caused by iron, folate, or Chapters 14 and 18 describe the interpretation of reticulocyte
vitamin B12 deficiency. An assessment of erythropoiesis can indices in patients with anemia.
determine the effectiveness of the bone marrow response,
however, and should be factored into the differential diagnosis Bone Marrow Examination
(see Differential Diagnosis). Bone marrow examination is usually not necessary to diagnose
hemolytic anemia. If conducted, however, bone marrow exam-
Complete Blood Count and Morphologic Features ination will reveal erythroid hyperplasia. The myeloid-to-
Peripheral blood film evaluation is crucial. An increase in poly- erythroid ratio decreases in hemolytic disease from a normal
chromatic RBCs (reticulocytosis) and nucleated RBCs repre- of about 3:1 as the erythroid component (the denominator)
sents bone marrow compensation for hemolysis or blood loss. increases. (The myeloid-to-erythroid ratio is defined in Chapter
An increase in the mean cell volume (MCV) is usually seen 16.) As always, the cellularity of the bone marrow should be
with extreme compensatory reticulocytosis resulting from the determined on a core biopsy specimen, rather than an aspi-
larger, prematurely released shift reticulocytes. The increase rated specimen, for a more accurate judgment.
must be assessed by comparison with the value seen early in
hemolysis, before the shift reticulocytes have emerged. The Laboratory Tests to Determine Specific
MCV may not increase above the reference range but rather may Hemolytic Processes
be above the value that was normal for that patient. Exceptions A well-made blood film is helpful in determining the cause of
occur if the hemolytic condition itself involves smaller cells that hemolytic anemia. Certain abnormalities found on the film,
counter the increased volume of the reticulocytes. In hereditary such as spherocytes, elliptocytes, acanthocytes, burr cells, sickle
spherocytosis, microspherocytes are the cause of the anemia, cells, target cells, agglutination, erythrophagocytosis, or para-
and the MCV may be within the reference range even when sites, may help reveal the specific disorder causing the hemo-
larger shift reticulocytes are generatedhence the importance lysis (Table 22-4).
of a baseline value for comparison. In other circumstances, such Leukocytosis and thrombocytosis may accompany acute
as severe burns, numerous schistocytes or microspherocytes hemolytic anemia and are considered reactions to the hemo-
may outnumber the reticulocytes, so that the MCV remains low. lytic process. Conversely, conditions that directly cause leuko-
cytosis, such as sepsis, also might cause hemolysis. When there
Reticulocyte Count is thrombocytosis, the platelets are generally large, which
The reticulocyte count is the most commonly used test to detect results in an increased mean platelet volume. Low platelet
accelerated erythropoiesis and is useful for this purpose. An counts in association with other signs of hemolysis may
increased reticulocyte count and an increased reticulocyte pro- indicate a microangiopathic hemolysis, such as disseminated
duction index (see Chapter 14) support a diagnosis of anemia intravascular coagulopathy. Other tests dealing with specific
secondary to blood loss or destruction of RBCs. When hemo- entities are discussed in subsequent chapters, including the
lysis is severe enough to produce anemia, the reticulocyte direct antiglobulin test, osmotic fragility test, autohemolysis
count is expected to be substantially increased. The association test, Heinz body test, RBC enzyme studies, serologic tests, and
is so strong that if a patient has an elevated reticulocyte count, immunophenotyping.
a cause of hemolysis should be sought. The increase usually
correlates well with the severity of the hemolysis. Exceptions
DIFFERENTIAL DIAGNOSIS
occur during aplastic crises of hemolytic anemias and in some
immunohemolytic anemias with hypoplastic marrow, which The differential diagnosis of hemolytic anemias incorporates
suggests that the autoantibodies were directed against the several intersecting lines of deduction. The first is to establish
bone marrow RBC precursors and circulating erythrocytes.10 the hemolytic nature of the anemia. A rapid decrease in
310 PART IV Hematopathology: Erythrocyte Disorders

hemoglobin concentration (e.g., 1g/dL per week) from previ-


ously normal levels can signal hemolysis when hemorrhage Whole blood
and hemodilution have been ruled out. Jaundice and reticulo- hemoglobin
cytosis provide additional confirmation of a hemolytic cause Plasma
hemoglobin
for an anemia of at least several days duration. When the
indirect fraction of the total serum bilirubin is elevated, hemo-
lytic jaundice is confirmed. An elevated urinary urobilinogen
level strengthens the conclusion. The rapid decrease in hemo-
Urinary
globin during an acute hemolytic episode, however, usually is hemoglobin Reticulocytes
apparent before reticulocytosis and bilirubinemia develop. For Plasma
acute hemolysis, hemoglobinemia and hemoglobinuria are Serum bilirubin
haptoglobin
expected with intravascular causes; therefore their absence sug-
gests an extravascular cause. RBC morphology and haptoglobin 1 2 3 4 5 6 7 8
levels can assist in differentiating an intravascular from an Hemolytic Time (days)
extravascular cause. Figure 22-5 is a graphic representation of event
A
the general timeline of the events in acute intravascular and
extravascular hemolysis. In chronic hemolysis, persistence of
hemoglobinemia, hemoglobinuria, decreased serum haptoglo-
bin level, indirect bilirubinemia, and elevated reticulocyte
count is expected, depending on the cause of the hemolysis.
Hemolytic anemias must be differentiated from other Whole blood
hemoglobin
anemias associated with bilirubinemia, reticulocytosis, or
both. Anemia with reticulocytosis but without bilirubinemia is
expected during recovery from hemorrhage not treated with
transfusion or with effective treatment of deficiencies such as
iron deficiency. Anemia that results from hemorrhage into a
body cavity is characterized by reticulocytosis during recovery Reticulocytes
and bilirubinemia due to catabolism of the hemoglobin in Serum haptoglobin Plasma
the hemorrhaged cells. The RBC morphology should remain bilirubin
normal throughout the event. Anemias associated with ineffec-
tive erythropoiesis, such as megaloblastic anemia, are essen- 1 2 3 4 5 6 7 8
tially hemolytic, with the cell death occurring in the bone Time (days)
Hemolytic
marrow. Bilirubinemia and elevated serum lactate dehydroge- B event
nase levels are to be expected, but the reticulocyte count is low.
Because most of the RBCs never reach the periphery, such Figure 22-5 A, Intravascular (fragmentation) hemolysis timeline. B, Extra
anemias are typically classified as anemias of diminished pro- vascular (macrophage-mediated) hemolysis timeline.
duction rather than as hemolytic anemias. As summarized in
Table 22-5, the differential diagnosis in each of these instances
may rely on negative results on tests for increased cell destruc-
tion or accelerated production.

TABLE 22-5 Differential Diagnosis of Hemolytic Anemias versus Other Causes of Indirect Bilirubinemia and Reticulocytosis
Hemoglobin Indirect Spherocytes or
Level Bilirubinemia Reticulocytosis Schistocytes
Hemolytic anemiaacute, intravascular Rapidly dropping Delayed Delayed Schistocytes
Hemolytic anemiaacute, extravascular Rapidly dropping Delayed Delayed Spherocytes
Hemolytic anemiachronic, intravascular Persistently low Persistent Persistent Schistocytes
Hemolytic anemiachronic, extravascular Persistently low Persistent Persistent Spherocytes
Acute hemorrhage Rapidly dropping Absent Absent Absent
Hemodilution Rapidly dropping Absent Absent Absent
Recovery from hemorrhage Rising Absent Present Absent
Treated anemia (iron deficiency) Rising Absent Present Absent
Hemorrhage into a body cavity Rapidly dropping Delayed Delayed Absent
Ineffective erythropoiesis (e.g., megaloblastic anemia) Dropping Persistent Absent Absent
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 311

SUMMARY
A hemolytic disorder is a condition in which there is increased Jaundice results from increased serum unconjugated bilirubin and
destruction of erythrocytes and a compensatory acceleration in is associated with all hemolytic anemias.
erythrocyte production by the bone marrow. The major clinical features of chronic inherited hemolytic
A hemolytic anemia develops when the bone marrow is unable to anemia are varying degrees of anemia, jaundice, the occurrence
compensate for the shortened survival of the RBCs. of crises, splenomegaly, and the development of cholelithiasis. In
Hemolytic anemias can be classified as acute or chronic, intravas- children, bone abnormalities develop as a result of accelerated
cular or extravascular, acquired or inherited, and intrinsic or erythropoiesis.
extrinsic. Laboratory studies providing evidence of hemolytic anemia include
Normally, most erythrocyte catabolism occurs extravascularly in tests for increased erythrocyte destruction and compensatory
the macrophages of the spleen, liver, and bone marrow. A small increase in the rate of erythropoiesis. The reticulocyte count is the
amount occurs intravascularly secondary to mechanical trauma. most commonly used laboratory test to identify accelerated eryth-
Hemoglobin from the RBCs is converted to heme and globin within ropoiesis. Elevated serum indirect (unconjugated) bilirubin level,
macrophages. Heme is further degraded to iron, carbon monoxide, with a normal serum direct (conjugated) bilirubin level and a
and unconjugated bilirubin. The bilirubin is secreted into the decreased haptoglobin level, indicate accelerated RBC destruc-
plasma, where it binds to albumin and is transported to the liver. tion. Other tests that are specific to a particular diagnosis also may
In the liver, the bilirubin is conjugated with glucuronic acid, excreted be needed.
as bile into the intestines, and converted to urobilinogen. Some The peripheral blood film evaluation can identify morphologic
urobilinogen is reabsorbed into the portal circulation and reexcreted changes that point to an intravascular or extravascular etiology
through the liver. A small amount of urobilinogen remains in the and to specific anemias. It is a valuable assessment in the dif-
plasma, and it is excreted by the kidney into the urine. ferential diagnosis of anemia.
In extravascular (macrophage-mediated) hemolytic anemia, there Hemolytic anemias must be differentiated from other anemias with
is an increase in unconjugated bilirubin in the plasma and an reticulocytosis, including the posthemorrhage state and recovery
increase in urobilinogen in the stool and urine. Spherocytes may from iron deficiency.
be seen on the blood film.
Signs of intravascular hemolysis include hemoglobinemia, (met) Now that you have completed this chapter, go back and
hemoglobinuria, and hemosiderinuria. Serum haptoglobin is read again the case study at the beginning and respond
markedly decreased or absent. Schistocytes may be seen on the to the questions presented.
blood film.

R E V I E W Q UESTIONS
1. The term hemolytic disorder in general refers to a disorder 4. An elderly white woman is evaluated for worsening
in which there is: anemia, with a decrease of approximately 0.5mg/dL of
a. Increased destruction of RBCs after they enter the hemoglobin each week. The patient is pale, and her skin
bloodstream and eyes are slightly yellow. She complains of extreme
b. Excessive loss of RBCs from the body fatigue and is unable to complete the tasks of daily living
c. Inadequate RBC production by the bone marrow without napping in mid-morning and mid-afternoon. She
d. Increased plasma volume with unchanged red cell mass also tires with exertion, finding it difficult to climb even
five stairs. Which of the features of this description points
2. RBC destruction that occurs when macrophages ingest and to a hemolytic cause for her anemia?
destroy RBCs is termed: a. Pallor
a. Extracellular b. Yellow skin and eyes
b. Extravascular c. Rate of decline of hemoglobin level
c. Intraorgan d. Tiredness on exertion
d. Extrahematopoietic
5. Which of the following tests provides a good indication of
3. Signs of hemolysis that are typically associated with intra- accelerated erythropoiesis?
vascular hemolysis only (and not extravascular hemolysis) a. Urine urobilinogen level
include all of the following except: b. Hemosiderin level
a. Hemoglobinuria c. Reticulocyte count
b. Hemosiderinuria d. Glycated hemoglobin level
c. Hemoglobinemia
d. Elevated urinary urobilinogen level
312 PART IV Hematopathology: Erythrocyte Disorders

6. A 5-year-old girl was seen by her physician several days 9. A patient has anemia that has been worsening over the last
ago and was diagnosed with pneumonia. Her mother has several months. The hemoglobin level has been declining
brought her to the physician again because the girls urine slowly with a drop of 1.5g/dL of hemoglobin over about
began to darken after the first visit and now is alarmingly 6 weeks. Polychromasia and anisocytosis are seen on the
dark. The girl has no history of anemia, and there is no blood film, consistent with the elevated reticulocyte count
family history of any hematologic disorder. The CBC and RBC distribution width (RDW). Serum levels of total
shows a mild anemia, polychromasia, and a few schisto- bilirubin and indirect fractions are normal. Urinary urobi-
cytes. This anemia could be categorized as: linogen level also is normal. When these findings are
a. Acquired, intravascular evaluated, the conclusion is drawn that the anemia does
b. Acquired, extravascular not have a hemolytic component. Based on the data given
c. Hereditary, intravascular here, why was hemolysis ruled out as the cause of the
d. Hereditary, extravascular anemia?
a. The decline in hemoglobin is too gradual to be
7. A patient has a personal and family history of a mild associated with hemolysis.
hemolytic anemia. The patient has consistently elevated b. The elevation of the reticulocyte count suggests a
levels of total and indirect serum bilirubin and urinary malignant cause.
urobilinogen. The serum haptoglobin level is consistently c. Evidence of increased protoporphyrin catabolism is
decreased, whereas the reticulocyte count is elevated. The lacking.
latter can be seen as polychromasia on the patients blood d. Elevated RDW points to an anemia of decreased
film along with spherocytes. Which of the findings reported production.
for this patient is inconsistent with a diagnosis of intravas-
cular hemolysis? 10. Which of the following sets of test results is typically
a. Elevated total and indirect serum bilirubin expected with intravascular hemolysis?
b. Elevated urinary urobilinogen
Serum Urine Urine Sediment
c. Decreased haptoglobin
Haptoglobin Hemoglobin Prussian Blue Stain
d. Spherocytes on the peripheral film
a. Increased Positive Positive
8. Select the statement that is true about bilirubin b. Decreased Negative Negative
metabolism. c. Decreased Positive Positive
a. Indirect bilirubin is formed in the liver by the d. Increased Positive Negative
addition of two sugar molecules to direct bilirubin.
b. Macrophages of the spleen liberate bilirubin during
hemoglobin catabolism.
c. Urobilinogen is not water soluble and is not excreted
in the urine.
d. Normally, the major fraction of bilirubin in the blood
is the direct (conjugated) form released from
macrophages.

REFERENCES
1. Crosby WH, Akeroyd JH: The limit of hemoglobin synthesis 7. Morgan WT, Liem HH, Sutor RP, et al: Transfer of heme
in hereditary hemolytic anemia. Am J Med 13:273-283, from heme-albumin to hemopexin. Biochem Biophys Acta
1952. 444:435-445, 1976.
2. Cui Y, Konig J, Leier I, et al: Hepatic uptake of bilirubin and 8. Pimstone N: Renal degradation of hemoglobin. Semin
its conjugates by the human organic anion transporter Hematol 9:31-42, 1972.
SLC21A6. J Biol Chem 276:9626-9630, 2001. 9. Sears DA, Anderson PR, Foy AL, et al: Urinary iron excretion
3. Glader B: Destruction of erythrocytes. In Greer JP, Foerster JL, and renal metabolism of hemoglobin in hemolytic disease.
Rodgers GH, et al, editors: Wintrobes clinical hematology, Blood 28:708-725, 1966.
ed 12, Philadelphia, 2009, Wolters Kluwer Health/Lippincott 10. Means RT Jr, Glader B: Anemia: general considerations. In
Williams & Wilkins, pp 159-169. Greer JP, Foerster JL, Rodgers GM, et al, editors: Wintrobes
4. Kristiansen M, Graversen JH, Jacobsen C, et al: Identification clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
of the haemoglobin scavenger receptor. Nature 409:198-201, Health/Lippincott Williams & Wilkins, pp. 779-809.
2001. 11. Crosby WH, Dameshek W: The significance of hemoglobine-
5. Tolosano E, Altruda F: Hemopexin: structure, function, and mia and associated hemosiderinuria, with particular refer-
regulation. DNA Cell Biol 21:297-306, 2002. ence to various types of hemolytic anemia. J Lab Clin Med
6. Delanghe JR, Langlois MR: Hemopexin: a review of biological 38:829-841, 1951.
aspects and the role in laboratory medicine. Clin Chim Acta 12. Fairley NH: Methemalbumin: clinical aspects. QJM 10:115-
312:13-23, 2001. 138, 1941.
CHAPTER 22 Introduction to Increased Destruction of Erythrocytes 313
13. Muller-Eberhard U: Hemopexin. N Engl J Med 283:1090- 17. Coburn RF: Endogenous carbon monoxide production in
1094, 1970. man. N Engl J Med 282:207-209, 1970.
14. Rosen H, Sears DA, Meisenzahl D: Spectral properties of 18. Karliner JS: Clinical usefulness and limitations of serum lactic
hemopexin-heme: the Schumm test. J Lab Clin Med 74:941- dehydrogenase determinations. In Griffiths JC, editor: Clinical
945, 1969. enzymology, New York, 1979, Masson, pp 25-29.
15. Trotman BW: Pigment gallstone disease. Gastroenterol Clin 19. Panzer S, Kronik G, Lechner K, et al: Glycosylated hemoglobin
North Am 20:111-126, 1991. (GHb): an index of red cell survival. Blood 59:1348-1350, 1982.
16. Marchand A, Galen RS, Van Lente F: The predictive value of 20. International Committee for Standardization in Haemato
serum haptoglobin in hemolytic disease. JAMA 243:1909- logy: Recommended method for radioisotope red cell sur-
1911, 1980. vival studies. Br J Haematol 45:659-666, 1980.
23 Increased
Intrinsic Defects Leading to
Erythrocyte Destruction
Elaine M. Keohane

OUTLINE OBJECTIVES
Red Blood Cell Membrane After completion of this chapter, the reader will be able to:
Abnormalities
Red Blood Cell Membrane 1. Describe the intrinsic cell properties that affect red 7. Describe the causes and pathophysiology of heredi-
Structure and Function blood cell (RBC) deformability. tary and acquired conditions characterized by
Hereditary Red Blood 2. Explain how defects in vertical and horizontal mem- acanthocytosis.
Cell Membrane brane protein interactions can result in a hemolytic 8. Describe the cause, pathophysiology, clinical mani-
Abnormalities anemia. festations, laboratory findings, and treatment for
Acquired Membrane 3. Compare and contrast the inheritance pattern, mem- paroxysmal nocturnal hemoglobinuria.
Defect: Paroxysmal brane proteins mutated, mechanism of hemolysis, 9. Compare and contrast the inheritance pattern,
Nocturnal typical RBC morphology, and clinical and laboratory pathophysiology, clinical symptoms, and typical
Hemoglobinuria findings of hereditary spherocytosis, hereditary laboratory findings of glucose-6-phosphate dehydro-
Red Blood Cell
elliptocytosis, and hereditary ovalocytosis. genase deficiency and pyruvate kinase deficiency.
Enzymopathies
4. Compare and contrast the RBC morphology and 10. Given the history, symptoms, laboratory findings,
Glucose-6-Phosphate
Dehydrogenase laboratory findings of hereditary spherocytosis and and a representative microscopic field from a periph-
Deficiency immune-associated hemolytic anemias. eral blood film for a patient with a suspected intrinsic
Pyruvate Kinase 5. Explain the principle, interpretation, and limitations hemolytic anemia, discuss possible causes of the
Deficiency of the osmotic fragility test in the diagnosis of heredi- anemia and indicate the data that support these
Other Enzymopathies tary spherocytosis. conclusions.
6. Describe the causes, pathophysiology, RBC morpho
logy, and clinical and laboratory findings of hemolytic
anemias characterized by stomatocytosis.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following
case study:

A 55-year-old man sought medical attention for the onset of chest pain. Physical examination
revealed slight jaundice and splenomegaly. The medical history included gallstones, and there was
a family history of anemia. A CBC yielded the following results:
Patient Results Reference Range
9
WBCs (10 /L) 13.4 4.5-11.5
RBCs (1012/L) 4.20 4.60-6.00
Hb (g/dL) 11.9 14.0-18.0
Hct (%) 32.4 40-54
MCV (fL) 77 80-100
MCH (pg) 28.3 26-32
MCHC (g/dL) 36.7 32-36
RDW (%) 22.9 11.5-14.5
Platelets (109/L) 290 150-450
Continued

314
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 315

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

The peripheral blood film revealed anisocytosis, polychroma-


sia, and spherocytes (Figure 23-1).
1. From the data given, what is a likely cause of the anemia?
2. What additional laboratory tests would be of value in
establishing the diagnosis, and what abnormalities in the
results of these tests would be expected to confirm your
impression?
3. What is the cause of this type of anemia?

Figure 23-1 Peripheral blood film for the patient in the case study
(500). (From Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
Philadelphia, 2009, Saunders.)

I
ntrinsic hemolytic anemias comprise a large group of dis- bilayer remains intact because transmembrane proteins
orders in which defects in the red blood cells (RBCs) embedded in the membrane anchor it to a two-dimensional
themselves result in premature hemolysis and anemia. protein lattice (skeleton) immediately beneath its surface.1
Intrinsic disorders can be divided into abnormalities of the Together, the transmembrane proteins and underlying skele-
RBC membrane, metabolic enzymes, or hemoglobin (Hb). ton provide structural integrity, cohesion, and mechanical sta-
Most of these defects are hereditary. This chapter covers defects bility to the cell.
in the RBC membrane and enzymes causing hemolytic anemia. In a normal life span of 120 days, RBCs must repeatedly
Chapter 26 covers qualitative hemoglobin disorders, and maneuver through very narrow capillaries and squeeze through
Chapter 27 covers quantitative hemoglobin disorders. the splenic sieve (narrow slits or fenestrations in the endothe-
lial cell lining of the splenic sinuses) as they move from the
splenic cords to the sinuses. To accomplish this without pre-
RED BLOOD CELL
mature lysis, the RBCs must have deformability, or the ability to
MEMBRANE ABNORMALITIES
repeatedly bend, stretch, distort, and then return to the normal
Red Blood Cell Membrane Structure discoid, biconcave shape.2 The cellular properties that enable
and Function RBC deformability include (1) the RBCs biconcave, discoid
The RBC maintains a biconcave discoid shape that is essential geometry, (2) the viscoelastic nature (elasticity) of its mem-
for normal function and survival for 120 days in the peripheral brane, and (3) its cytoplasmic viscosity (see Chapter 9).3,4
circulation. The key to maintaining this shape is the plasma The biconcave, discoid geometry of the RBC is dependent
membrane, a lipid bilayer embedded with proteins and con- on vertical and horizontal interactions between the transmem-
nected to an underlying protein skeleton (see Figure 9-2). The brane and skeletal proteins (see Tables 9-5 and 9-6).3 Two
insoluble lipid portion serves as a barrier to separate the vastly transmembrane protein complexes, the ankyrin complex and
different ion and metabolite concentrations of the interior of protein 4.1 junctional complex, provide vertical structural
the RBC from its external environment, the blood plasma. The integrity to the cell by anchoring the lipid bilayer to the under-
concentration of the constituents in the cytoplasm is tightly lying spectrin skeleton.3,5 In the ankyrin complex, ankyrin
regulated by proteins embedded in the membrane that serve and protein 4.2 link transmembrane proteins, band 3, and
as pumps and channels for movement of ions and other mate- RhAG (the Rh-associated glycoprotein) to the skeleton.6,7 In the
rial between the RBCs interior and the blood plasma. Various protein 4.1 junctional complex, protein 4.1R links transmem-
membrane proteins also act as receptors, RBC antigens, brane proteins, glycophorin C, and XK, Rh, and Duffy proteins
enzymes, and support for the surface carbohydrates to form a to the skeleton, and adducin and dematin link with transmem-
protective glycocalyx with the surface glycolipids. The lipid brane proteins, band 3, and glucose transport (glut 1) (see
316 PART IV Hematopathology: Erythrocyte Disorders

Figure 9-2).8 These interactions are called vertical because they


are perpendicular to the plane of the cytoskeleton. They prevent BOX 23-1 Classification of Major Hereditary
loss of membrane and the resultant decrease in the surface Membrane Defects Causing Hemolytic Anemia
areatovolume ratio of the RBC.5 Two major skeletal proteins,
-spectrin and -spectrin, interact laterally with each other to Mutations That Alter Membrane Structure
form antiparallel heterodimers, which link with other spectrin Hereditary spherocytosis
heterodimers to form tetramers. The spectrin heterodimers Hereditary elliptocytosis/pyropoikilocytosis
also form a spectrinactinprotein 4.1R junctional complex Hereditary ovalocytosis (Southeast Asian ovalocytosis)
with accessory proteins (tropomyosin, tropomodulin, dematin,
and adducin), thus linking the spectrin tetramers in a two- Mutations That Alter Membrane
dimensional lattice.9,10 These proteins provide horizontal Transport Proteins
mechanical stability, which prevents the membrane from frag- Dehydrated hereditary stomatocytosis
menting in response to mechanical stress.3,5 Overhydrated hereditary stomatocytosis
Factors that impact the elasticity of the cell are not as clear.
Interactions between the spectrin dimers and the junctional
complexes are flexible and allow for movement as the RBCs transmembrane proteins and the underlying protein cyto
stretch and bend (see Figure 9-4).10 In addition, the ability of skeleton. HS has worldwide distribution and affects 1 in 3000
spectrin repeats to unfold and refold is likely to be one of the individuals of northern European ancestry.10 In 75% of fami-
determinants of membrane elasticity.10,11 lies, it is inherited as an autosomal dominant trait and is
The cytoplasmic viscosity depends on the concentration of expressed in heterozygotes who have one affected parent.
hemoglobin, as well as the maintenance of the proper cell Homozygotes are rare; such patients present with severe hemo-
volume by the normal functioning of various channels and lytic anemia, but have asymptomatic parents.13 In approxi-
pumps that allow the passage of ions, water, and macromol- mately 25% of cases, the inheritance is nondominant, with
ecules in and out of the cell.3 some autosomal recessive cases.10,13
A defect in the RBCs that changes the membrane geometry, Pathophysiology. HS results from gene mutations in
its elasticity, or the viscosity of the cytoplasm affects RBC which the defective proteins disrupt the vertical linkages
deformability and can result in premature hemolysis and between the lipid bilayer and the skeletal network.5 Various
anemia.3 The RBC membrane is discussed in more detail in mutations in six known genes can result in the HS phenotype
Chapter 9; the reader is encouraged to review that chapter (see Table 23-1). Mutations can occur in genes for (1) anchor-
when studying defects in the membrane. ing proteins, including ANK1, which codes for ankyrin (40%
to 65% of cases in the United States and Europe; 5% to 10% of
Hereditary Red Blood Cell cases in Japan), and EPB42, which codes for protein 4.2 (fewer
Membrane Abnormalities than 5% of cases in the United States and Europe; 45% to 50%
Most defects in the RBC membrane that can cause hemolytic of cases in Japan); (2) transmembrane proteins, including
anemia are hereditary; however, acquired defects also exist. SLC4A1, which codes for band 3 (20% to 35% of cases), and
Hereditary RBC membrane defects have historically been clas- RHAG, which codes for RhAG (fewer than 1% of cases); or (3)
sified by morphologic features. The major disorders are heredi- skeletal proteins, including SPTA1, which codes for -spectrin
tary spherocytosis, characterized by spherocytes, and hereditary (fewer than 5% of cases) and SPTB, which codes for -spectrin
elliptocytosis, characterized by elliptical RBCs. Hereditary (15% to 30% of cases).10,14 Because of the vertical interactions
pyropoikilocytosis, a variant of hereditary elliptocytosis, is of the transmembrane proteins and skeletal protein lattice, a
characterized by marked poikilocytosis and heat sensitivity. primary mutation in one gene may have a secondary effect on
Other less common membrane disorders include hereditary another protein in the membrane. For example, primary muta-
ovalocytosis, overhydrated hereditary stomatocytosis, and tions in ANK1 result in both ankyrin and spectrin deficiencies
dehydrated hereditary stomatocytosis. Hereditary membrane in the RBC membrane.14,15 In approximately 10% of patients,
defects can also be classified as those that affect membrane no mutation is identified.10
structure (and alter geometry and elasticity) and those that The defects in vertical membrane protein interactions cause
affect membrane transport (and alter cytoplasmic viscosity) RBCs to lose unsupported lipid membrane over time because
(Box 23-1).10,12 The membrane structural defects can be further of local disconnections of the lipid bilayer and underlying
divided into those that affect vertical membrane protein inter- skeleton. Essentially, small portions of the membrane peel off
actions and those that affect horizontal membrane protein with little loss of volume. The RBCs acquire a decreased surface
interactions.5,10 The major hereditary membrane defects are areatovolume ratio, and the cells become spherical. These
described in Table 23-1. cells do not have the deformability of normal biconcave discoid
RBCs, and their survival in the spleen is decreased.4,13,14 As the
Mutations That Alter Membrane Structure spherocytes attempt to move through the narrow, elliptical
Hereditary Spherocytosis. Hereditary spherocytosis fenestrations of the endothelial cells lining the splenic sinu-
(HS) is a heterogenous group of hemolytic anemias caused by soids, they acquire further membrane loss or become trapped
defects in proteins that disrupt the vertical interactions between and are rapidly removed by the macrophages of the red pulp
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 317

TABLE 23-1 Characteristics of Major Hemolytic Anemias Caused by Hereditary Membrane Defects
Inheritance Deficient Protein Typical RBC Clinical Findings/
Condition Pattern (Mutated Gene) Pathophysiology Morphology Comments
Hereditary 75% autosomal -Spectrin (SPTA1) Defect in protein(s) that Spherocytes, Varies from
spherocytosis dominant -Spectrin (SPTB) disrupt vertical membrane polychromasia asymptomatic to
25% nondominant Band 3 (SLC4A1) interactions between severe
Rh-associated transmembrane proteins and Typical features:
protein (RHAG) underlying skeleton; loss of splenomegaly,
Ankyrin (ANK1) membrane and decreased jaundice, anemia
Protein 4.2 (EPB42) surface areatovolume
ratio
Hereditary Autosomal -Spectrin (SPTA1) Defect in proteins that disrupt Few to 100% 90% of cases
elliptocytosis dominant -Spectrin (SPTB) the horizontal linkages in the elliptocytes, asymptomatic; 10% of
Protein 4.1R (EPB41) protein skeleton; loss of schistocytes in cases show moderate
mechanical stability of severe cases to severe anemia
membrane
Hereditary Autosomal -Spectrin (SPTA1) Severe defect in spectrin that Elliptocytes, Severe anemia
pyropoikilocytosis recessive disrupts horizontal linkages schistocytes,
(subtype of in protein skeleton; severe microspherocytes
hereditary RBC fragmentation
elliptocytosis)
Hereditary Autosomal Band 3 (SLC4A1); Defect in band 3 causing 30% oval cells with Asymptomatic or mild
ovalocytosis, or dominant only one known increased membrane rigidity; one or two hemolysis, resistance
Southeast Asian mutation only exists in heterozygous transverse ridges to malaria; prevalent
ovalocytosis state in some areas of
Southeast Asia
Overhydrated Autosomal Rh-associated Increased membrane Stomatocytes Moderate to severe
hereditary dominant protein (RHAG) permeability to sodium and (5%-50%), hemolytic anemia;
stomatocytosis potassium; increased macrocytes splenectomy is
intracellular sodium causing contraindicated due to
influx of water, increase in thrombotic risk
cell volume, and decreased
cytoplasmic viscosity
Dehydrated Autosomal Unknown; mapped Increased membrane Target cells, burr Mild to moderate
hereditary dominant to 16q23-q24 in permeability to potassium; cells, stomatocytes anemia, splenomegaly;
stomatocytosis some patients decreased intracellular (<10%), RBCs may lead to fetal loss,
potassium resulting in loss with puddled hydrops fetalis; may
of water from cell, decrease hemoglobin at be accompanied by
in cell volume, and increased periphery pseudohyperkalemia
cytoplasmic viscosity

RBC, Red blood cell.

of the spleen.4,13 In addition, as the RBCs are sequestered in the results in a higher intracellular concentration of sodium and
spleen, the membrane can acquire yet more damage, lose more lower concentration of potassium compared with normal cells.
lipid membrane, and become more spherical due to splenic The abnormality is not related to defects in cation transport
conditioning.4,13,14 The conditioning may be enhanced by the proteins, and it occurs regardless of the type of primary muta-
acidic conditions in the spleen and the prolonged contact of tion causing the HS.16
the RBCs with macrophages.13,14 Low levels of adenosine tri- Clinical and laboratory findings. Symptomatic HS has
phosphate (ATP) and glucose, and phagocyte-produced free three key clinical manifestations: anemia, jaundice, and sple-
radicals, which cause oxidative damage, may also play a role nomegaly. Symptoms of HS may first appear in infancy, child-
(Figure 23-2).14 hood, or adulthood, or even at an advanced age. There is wide
In HS, RBC membranes also have abnormal permeability variation in symptoms. Silent carriers are clinically asymptom-
to cations, particularly sodium and potassium, which is likely atic with normal laboratory findings and usually are identified
due to disruption of the integrity of the protein skeleton.16 This only if they are the parents of a child with recessively inherited
318 PART IV Hematopathology: Erythrocyte Disorders

Reduced Poor
surface-to- cellular
volume ratio deformability
(spherocytosis)

Skeletal
membrane Membrane Membrane Splenic Splenic
skeletal instability loss conditioning trapping
defect

ATP
depletion

Acid-induced RBC stasis


damage Low glucose
Acid pH
Macrophage
Macrophage contact
processing

Oxidative
damage

Hemolysis
Phagocytosis
Osmotic lysis

Figure 23-2 Pathophysiology of splenic trapping and destruction of spherocytes. ATP, Adenosine triphosphate; RBC, Red blood cell. (Modified from Becker
PS, Lux SE: Disorders of the red cell membrane. In Nathan DG, Oski FA, editors: Hematology of infancy and childhood, ed 4, Philadelphia, 1993, Saunders,
pp 529-633.)

HS.4,13 Approximately 20% of patients have mild HS and filled with hemoglobin and lack a central pallor (Figure 23-3).
are asymptomatic, because an increase in erythropoiesis com- Normal-appearing RBCs, along with polychromasia and
pensates for the RBC loss.10,13 They usually have normal hemo- varying degrees of anisocytosis and poikilocytosis, are present.
globin levels, but may show subtle signs of HS, with an In addition to spherocytes, occasionally other RBC morpho-
increased reticulocyte count of up to 6% and a few spherocytes logic variants may be observed in some types of mutations:
on the peripheral blood film.4,10,15 Mild HS may first become acanthocytes in some -spectrin mutations,17 pincered or
evident during pregnancy, during illnesses that cause spleno- mushroom-shaped cells in some cases of band 3 deficiency in
megaly (such as infectious mononucleosis), or in aging when patients without splenectomy,18 ovalostomatocytes in homo-
the rate of erythropoiesis starts to decline.13,15 Approximately zygous EPB4.2 mutations,19 and stomatocytes in those with no
60% of patients have moderate HS with incompletely compen- expression of RHAG, the Rh-null phenotype.20 Note that sphe-
sated hemolysis, hemoglobin levels greater than 8 g/dL, reticu- rocytes are not specific for HS and can be seen in other heredi-
locyte counts greater than 6%, and spherocytes on the tary and acquired conditions.
peripheral blood film.10,14,15 Jaundice is seen at some time in Because of the spherocytosis, HS patients have an increase
about half of these patients, usually during viral infections. in the mean cell hemoglobin concentration (MCHC) to
About 10% of patients have moderately severe HS with hemo- between 35 and 38mg/dL and an increase in the red cell dis-
globin levels usually in the range of 6 to 8 g/dL and reticulocyte tribution width (RDW) to greater than 14%.4,15 Automated
counts greater than 10%.10,14 About 3% to 5% of patients hematology analyzers can detect the hyperdense RBCs found
have severe HS with hemoglobin levels below 6 g/dL and in HS.13 Biochemical evidence of extravascular hemolysis may
reticulocyte counts greater than 10%.10,14 Patients with severe be present in moderate to severe forms of HS, and the extent
HS are usually homozygous for HS mutations and require is dependent upon the severity of the hemolysis. This includes
regular transfusions.10 Splenomegaly is found in about half of a decrease in serum haptoglobin level and an increase in serum
young children and 75% to 95% of older children and adults indirect bilirubin level (see Chapter 22). The bone marrow
with HS.10 shows erythroid hyperplasia due to the increased demand for
The hallmark of HS is spherocytes on the peripheral blood RBCs to replace the circulating spherocytes that are prema-
film. When present in patients with childhood hemolytic turely destroyed, but bone marrow analysis is not required for
anemia and a family history of similar abnormalities, the diagnosis.
uniform spherocytes are highly suggestive of HS. Some of Special tests. In cases in which HS is suspected, but the
these are microspherocytessmall, round, dense RBCs that are family history and mode of inheritance are not clear, or there
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 319

tail

A Figure 23-4 Erythrocyte osmotic fragility curve. A, Curve in thalassemia


showing two cell populations: one with increased fragility (lower left of curve)
and one with decreased fragility (upper right of curve). B, Normal curve.
C, Curve indicating increased fragility, as in hereditary spherocytosis.

useful confirmatory test for HS; however, a major drawback is


its lack of sensitivity and specificity.15 In terms of sensitivity,
the test result can be normal in patients with HS, especially the
mild forms, so a normal osmotic fragility test finding does not
rule out HS.15 In patients with mild HS and a normal osmotic
fragility test result, incubating the blood at 37 C for 24 hours
before performing the test may enable its detection of mild
disease. During this incubation period, HS cells tend to become
more spherical. Patients who have increased osmotic fragility
only when their blood is incubated tend to have a low number
B of spherocytes in the total RBC population. In the osmotic
fragility test without incubation, the existence of a distinct
Figure 23-3 Spherocytes (peripheral blood, 1000). subpopulation of the most fragile cells, those most condi-
tioned by the spleen, is reflected by the presence of a tail on
the osmotic fragility curve (see Figure 23-4).13 After splenec-
are atypical clinical and/or laboratory findings, further special tomy is performed, the osmotic fragility improves, and this
testing is needed for diagnosis.15 subpopulation of conditioned cells disappears.13 The osmotic
Osmotic fragility. The osmotic fragility test demonstrates fragility test is also nonspecific. A result indicating increased
increased RBC fragility in blood samples in which cells have fragility does not differentiate between HS and spherocytosis
decreased surface areatovolume ratios. In this test, when caused by other conditions, such as burns, immune hemolytic
RBCs are put into a series of increasingly hypotonic sodium anemias, and other acquired disorders.13,15
chloride (NaCl) solutions, water enters the cells until equilib- Autohemolysis test. The autohemolysis test is relatively
rium is achieved. As this phenomenon occurs, the cells swell sensitive to HS but is not routinely used in many laboratories
until the internal volume is too great, which causes them to because the test is time consuming. It probably has sensitivity
lyse. Because spherocytes already have a decreased surface similar to that of the osmotic fragility test with incubation.4,13
areatovolume ratio, they lyse in less hypotonic solutions When normal RBCs are incubated in their own plasma, hemo-
than normal-shaped, biconcave RBCs and thus have increased lysis gradually takes place. In HS, because of the loss of mem-
osmotic fragility. A plot of an osmotic fragility curve is shown brane and the inability to maintain the cation gradient, the
in Figure 23-4. Normal biconcave RBCs show initial hemolysis hemolysis is much greater. Normal samples generally have less
at 0.45% NaCl, and hemolysis is complete at 0.30% NaCl. If than 5.0% hemolysis at the end of the 48-hour incubation
the curve is shifted to the left, the patients RBCs have increased period, and they have less than 1.0% if a source of glucose is
osmotic fragility. Conversely, if the curve is shifted to the right, added. The glucose provides energy to drive the cation pumps
the RBCs have decreased osmotic fragility. Decreased osmotic to help maintain the cells. In HS, the hemolysis is 10% to 50%,
fragility is found in conditions characterized by numerous which corrects considerably but not to the normal range
target cells, such as thalassemia. The osmotic fragility test is a when glucose is added. In an HS patient with numerous
320 PART IV Hematopathology: Erythrocyte Disorders

microspherocytes conditioned in vivo by the spleen, the hemo- develop folic acid deficiency resulting from increased folate
lysis may not correct with glucose. utilization to support the chronic erythroid hyperplasia in the
Other tests. In atypical HS cases additional tests may bone marrow.13 This phenomenon is termed megaloblastic crisis
be required to identify the primary gene defect or to deter- and is particularly acute during pregnancy and during recovery
mine the inheritance pattern.15 Sodium dodecyl sulfate- from an aplastic crisis. Providing folic acid supplementation to
polyacrylamide gel electrophoresis (SDS-PAGE) can be used to patients with moderate and severe HS avoids this complica-
identify membrane protein deficiencies by electrophoretic sep- tion.15 About half of patients, even those with mild disease, also
aration of the various proteins in solubilized RBC membranes experience cholelithiasis (bilirubin stones in the gallbladder
with quantitation of the proteins by densitometry.13,15 Mem- or bile ducts) due to the chronic hemolysis.13 Chronic ulcer-
brane proteins can also be quantitated by radioimmunoassay.13 ation or dermatitis of the legs is a rare complication.13
Variation in membrane surface area and cell water content can Treatment. Mild HS usually requires no treatment.15 In
be determined by osmotic gradient ektacytometry.21 The ekta- moderate and severe cases, splenectomy prevents clinically sig-
cytometer is a laser-diffraction viscometer that records the laser nificant hemolysis, results in longer RBC survival in the peri
diffraction pattern of a suspension of RBCs exposed to constant pheral blood, and helps prevent gallstones by decreasing the
shear stress in solutions of varying osmolality from hypotonic amount of hemolysis and thus the amount of bilirubin pro-
to hypertonic. An RBC osmotic deformability index is calcu- duced.15 Patients with uncomplicated disease have an excellent
lated and plotted against the osmolality of the suspending response. The major drawback of splenectomy is the lifelong
solution to generate an osmotic gradient deformability profile.21 risk of overwhelming sepsis and death, particularly after infec-
This method, however, is available only in specialized labora- tion with Streptococcus pneumoniae.13,15 Patients receive pneu-
tories.12,13 Patient blood can be screened for HS after incuba- mococcal vaccination and penicillin prophylaxis to reduce the
tion with eosin-5-maleimide (EMA) and measurement of the risk of sepsis, but this does not completely eliminate the risk.15
fluorescence in a flow cytometer. EMA binds to band 3 and Because infants and young children are especially susceptible
Rh-related proteins in the RBC membrane, and RBCs from to postsplenectomy sepsis, splenectomy is usually postponed
HS patients with deficiencies in these proteins have 25% to until after age 6 years.15 In a recent nationwide sample of 1657
30% lower fluorescence than RBCs from normal controls and children (aged 5 to 12 years) with HS who underwent splenec-
from patients with spherocytes due to immune-mediated tomy, there were no cases of postoperative sepsis and no fatali-
hemolysis.22 ties from any cause during hospitalization for the surgery.24
The hypertonic cryohemolysis test is based on the fact that More recently, partial splenectomy has been performed in
cells from HS patients are particularly sensitive to cooling at young children with severe HS, but further evaluation of the
0 C in hypertonic solutions.23 The percentage of hemolysis is risks and benefits of this procedure are needed.13
calculated after the patients RBCs are incubated in buffered After splenectomy, spherocytes are still apparent on the
0.7mol/L sucrose, first at 37 C for 10 minutes, then at 0 C blood film and the osmotic fragility is still increased, but
for 10 minutes. Normal cells show 3% to 15% hemolysis, conditioned microspherocytes are no longer present, as evi-
whereas RBCs in HS have greater than 20% hemolysis.23 This denced by the disappearance of the tail of the osmotic fragility
test is sensitive in detecting HS, but increased hemolysis can also curve. All of the typical changes in RBC morphology seen
occur in Southeast Asian ovalocytosis, some types of hereditary after splenectomy also are observed, including Howell-Jolly
elliptocytosis, and congenital dyserythropoietic anemia type bodies, target cells, and Pappenheimer bodies (Figure 23-5).
II.15,23 Molecular techniques for detection of genetic mutations
can be difficult and costly to use because of the large size of
the genes involved and the number of possible mutations.12,13
The polymerase chain reaction procedure followed by single-
strand conformational polymorphism analysis can identify
potential regions in HS genes that may contain a mutation.13,15
Once a region has been identified, nucleic acid sequencing can
be performed to identify the specific mutation.13
Complications. Although most patients with HS have a
well-compensated hemolysis and are rarely symptomatic, com-
plications may occur that require medical intervention. Patients
may experience various crises, classified as hemolytic, aplastic,
and megaloblastic.13 Hemolytic crises are rare and usually asso-
ciated with viral syndromes. In aplastic crises there is a dramatic
decrease in hemoglobin level and reticulocyte count. The crisis
usually occurs in conjunction with parvovirus B19 infection,
which suppresses erythropoiesis, and patients can become
rapidly and severely anemic, often requiring transfusion.15 This Figure 23-5 Red blood cell morphology in hereditary spherocytosis after
complication is more common in children, but can occur in splenectomy. Howell-Jolly bodies and spherocytes are seen (peripheral
adults.13,15 Patients with moderate and severe HS can also blood, 1000).
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 321

Reticulocyte counts decrease to high-normal levels, and the Hereditary Elliptocytosis. Hereditary elliptocytosis
anemia is usually corrected. Leukocytosis and thrombocytosis (HE) is a heterogenous group of hemolytic anemias caused by
are present. Bilirubin levels decrease but may remain in the defects in proteins that disrupt the horizontal or lateral interac-
high-reference range. tions in the protein cytoskeleton. It reportedly exists in all of
Occasionally, a patient does not improve because of an its forms in 1 in 2000 to 1 in 4000 individuals, but because the
accessory spleen missed during surgery or the accidental auto- majority of cases are asymptomatic, the actual incidence is not
transplantation of splenic tissue during splenectomy, and known.25 The disease is more common in Africa and Mediter-
hemolysis may resume years later.13 The resumption of splenic ranean regions where there is a high prevalence of malaria. The
function can be ascertained from liver or spleen scans.13 prevalence in West Africa of certain spectrin mutations associ-
Patients with severe HS usually require regular transfusions. ated with HE is between 0.6% and 1.6%.26 The molecular basis
Transfusions are rarely needed in the less severe forms of HS.14 for the association of spherocytosis and malaria is unknown.
Differential diagnosis. HS must be distinguished from The inheritance pattern in HE is autosomal dominant.10
immune-related hemolytic anemia with spherocytes. Family Pathophysiology. HE results from gene mutations in
history and evaluation of family members, including parents, which the defective proteins disrupt the horizontal linkages in
siblings, and children of the patient, help differentiate the the protein cytoskeleton and weaken the mechanical stability
hereditary disease from the acquired disorder. The immune of the membrane.5 The HE phenotype can result from various
disorders with spherocytes are usually characterized by a posi- mutations in at least three genes: SPTA1, which codes for
tive result on the direct antiglobulin test (DAT), whereas the -spectrin (65% of cases); SPTB, which codes for -spectrin
results are negative in HS. The osmotic fragility test is not (30% of cases); and EPB41, which codes for protein 4.1R (5%
diagnostic of HS, because the cells in acquired hemolytic of cases) (see Table 23-1).10,25 The spectrin mutations disrupt
anemia with spherocytes also show increased osmotic fragility. spectrin dimer interactions and the EPB41 mutations result in
In cases with a family history of HS, typical physical findings, weakened spectrinactinprotein 4.1R junctional complexes.10
an increased reticulocyte count, spherocytes on the peripheral RBCs are biconcave and discoid at first, but become elliptical
blood film, and a negative result on the DAT, no further special over time after repeated exposure to the shear stresses in the
testing is needed for the diagnosis of HS.15 The typical clinical peripheral circulation. The extent of the disruption of the spec-
and laboratory findings in HS are summarized in Table 23-2. trin dimer interactions seems to be associated with the severity
of the clinical manifestations.10 In severe cases, the protein
skeleton is weakened to such a point that cell fragmentation
occurs. As a result, there is membrane loss and a decrease in
TABLE 23-2 Typical Clinical and Laboratory Findings surface areatovolume ratio that reduces the deformability of
in Hereditary Spherocytosis (HS) the RBCs. The damaged RBCs become trapped or acquire
further damage in the spleen, which results in extravascular
Clinical manifestations Splenomegaly
Anemia* hemolysis and anemia. In general, patients who are heterozy-
Jaundice (can be intermittent) gous for a mutation are asymptomatic and their RBCs have a
Mode of inheritance 75% autosomal dominant normal life span; those who are homozygous for a mutation
25% nondominant or compound heterozygous for two mutations can have
Complete blood count results Hemoglobin decreased* moderate to severe anemia that can be life-threatening.10
Mean cell hemoglobin concentration The RBCs in HE patients all show some degree of decreased
increased thermal stability. Cases in which the RBCs show marked RBC
Red cell distribution width increased fragmentation upon heating were previously classified as
Reticulocyte count increased* hereditary pyropoikilocytosis (HPP). HPP is now considered a
Peripheral blood film findings Spherocytes severe form of HE that exists in either the homozygous or
Polychromasia compound heterozygous state.10
Direct antiglobulin test result Negative Patients with the Leach phenotype lack the Gerbich anti-
Indicators of hemolysis Increased serum indirect bilirubin gens, glycoprotein C (GPC), and glycoprotein D (GPD).4 The
Decreased serum haptoglobin phenotype is due to a mutation in the genes for GPC and GPD
Osmotic fragility test Usually increased and results in the absence of the glycoproteins in the RBC
Special tests Not required for diagnosis of HS with membrane.27 The Gerbich antigens are normally expressed on
the typical features listed above the extracellular domains of GPC and GPD and thus are absent
in this condition. Heterozygotes have normal RBC morpho
Adapted from Botlon-Maggs PHB, Stevens RF, Dodd NJ, etal: Guidelines for the logy, and homozygotes have mild elliptocytosis but no
diagnosis and management of hereditary spherocytosis, Br J Haematol 126:459, anemia.4 The reason for the elliptocyte morphology may be a
2004.
*Varies with severity of HS and ability of the bone marrow to compensate for the defect in the interaction between GPC and protein 4.1R in the
hemolysis. junctional complex.4

With some rare mutations, acanthocytes, pincered cells, stomatocytes, or ovalocytes Clinical and laboratory findings. The vast majority of
may be seen in addition to spherocytes.

Incubation of blood at 37 C for 24 hours prior to testing may be required to detect patients with HE are asymptomatic, and only about 10%
mild disease. A normal osmotic fragility test finding does not rule out HS. have moderate to severe anemia.10 Some may have a mild
322 PART IV Hematopathology: Erythrocyte Disorders

Figure 23-6 Red blood cell morphology in hereditary elliptocytosis


(peripheral blood, 1000). (From Carr JH, Rodak BF: Clinical hematology
atlas, ed 3, Philadelphia, 2009, Saunders.)

compensated hemolysis as evidenced by a slight increase in the


reticulocyte count and a decrease in haptoglobin level, or
develop transient hemolysis in response to other conditions
such as viral infections and pregnancy. Often an asymptomatic
patient is diagnosed after a peripheral blood film is examined
for another condition. Rarely, heterozygous parents with undi- B
agnosed, asymptomatic HE have offspring who are homozy- Figure 23-7 A, Morphology of red blood cells in hereditary pyropoikilo-
gous or compound heterozygous for their mutation(s) and cytosis before incubation. B, Morphology after 1 hour at 45 C (peripheral
have moderate to very severe hemolysis. blood, 500).
The characteristic finding in HE is elliptical or cigar-shaped
RBCs on the peripheral blood film in numbers that can vary
from a few to 100%4 (Figure 23-6). The number of elliptocytes Mutation screening using molecular tests or quantitation of
does not correlate with disease severity.4 Investigation of ellip- membrane proteins by SDS-PAGE may be also be performed.
tocytosis begins by taking a thorough patient and family As in HS, patients with moderate or severe hemolytic
history and performing a physical examination, and examining anemia due to HE can develop cholelithiasis due to bilirubin
the peripheral blood films of the parents.4 Other laboratory gallstones, and hemolytic, aplastic, and megaloblastic crises
tests may be needed to rule out other conditions in which can occur (see earlier section on HS complications).
elliptocytes may be present, such as iron deficiency anemia, Treatment. Asymptomatic HE patients require no treat-
thalassemia, megaloblastic anemia, myelodysplastic syndrome, ment. All HE patients who are significantly anemic and show
and primary myelofibrosis.4 In the homozygous or compound signs of hemolysis respond well to splenectomy.4 Transfusions
heterozygous states, the anemia is moderate to severe, the are occasionally needed for life-threatening anemia.
osmotic fragility is increased, and biochemical evidence of
excessive hemolysis is present. The peripheral blood film in Hereditary Ovalocytosis (Southeast Asian Ovalocyto-
patients with the HPP phenotype shows extreme poikilocytosis sis). Hereditary ovalocytosis or Southeast Asian ovalocytosis
with fragmentation, microspherocytosis, and elliptocytosis (SAO) is a condition caused by a mutation in the gene for band
similar to that in patients with thermal burns (Figure 23-7, A). 3 that results in increased rigidity of the membrane and resis-
The mean cell volume (MCV) is very low (50 to 75 fL) because tance to invasion by malaria.3 It is common in the malaria belt
of the RBC fragments.4 RBCs in the HPP phenotype show of Southeast Asia, where its prevalence can reach 30%.29 The
marked thermal sensitivity. After incubation of a blood sample inheritance pattern is autosomal dominant, and all patients
at 41 C to 45 C, the RBCs fragment (Figure 23-7, B).13 identified are heterozygous.3
Normal RBCs do not fragment until reaching a temperature of Pathophysiology. SAO is the result of one mutation, a
49 C. Thermal sensitivity is not specific for HPP, however; it deletion of 27 base pairs in SLC4A1, the gene that codes for
also occurs in cases of HE with spectrin mutations.4,13 RBCs band 3.30 Deletion occurs at the interface between the trans-
with the HPP phenotype show a lower fluorescence than RBCs membrane and cytoplasmic domains.31 The mutation causes
in HS when incubated with eosin-5-maleimide and analyzed an increase in membrane rigidity. There is tighter binding of
by flow cytometry (see earlier section on special tests for HS).28 band 3 to ankyrin and decreased lateral mobility of band 3 in
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 323

the membrane.13,29 RBC membranes also have decreased elas-


ticity as measured by ektacytometry and micropipette aspira-
tion.3,29 The molecular mechanism responsible for the increased
membrane rigidity that results from the band 3 mutation has
not yet been elucidated.3,4 These cells are resistant to cerebral
malaria caused by Plasmodium falciparum.32 The cells also have
an increased permeability to sodium and potassium that is
enhanced at cold temperatures.32
Clinical and laboratory findings. In patients with SAO,
hemolysis is mild or absent. On a peripheral blood film, typical
cells of SAO are oval RBCs with one to two transverse bars or
ridges and usually comprise about 30% of the RBCs.3,13 No
treatment is required for this condition.

Mutations That Alter Membrane Transport Proteins Figure 23-8 Red blood cell morphology in hereditary stomatocytosis
RBC volume is regulated by a number of membrane proteins (peripheral blood, 1000).
that serve as passive transporters, active transporters, and ion
channels. When RBCs lose the ability to regulate volume, the reticulocytosis, reduced erythrocyte potassium concentration,
cells are prematurely hemolyzed. Cell volume is determined elevated erythrocyte sodium concentration, and increased net
by the intracellular concentration of cations, particularly cation content in the erythrocytes.3 Because of the increased
sodium.10 If the total cation content is increased, water enters cell volume, the RBCs have increased osmotic fragility due to
the cell and increases the cell volume, forming a stomatocyte. a decreased surface areatovolume ratio.10 Splenectomy is
If the total cation content is decreased, water leaves the cell, contraindicated in OHSt, because it results in an increased risk
which decreases the cell volume and produces a dehydrated of thromboembolic complications.3
RBC, sometimes called a xerocyte.
Hereditary stomatocytosis comprises a group of heteroge- Dehydrated Hereditary Stomatocytosis. Dehydrated
nous conditions in which the RBC membrane leaks monova- hereditary stomatocytosis (DHSt) is an autosomal dominant
lent cations. The two major categories are overhydrated hemolytic anemia due to a defect in membrane cation perme-
hereditary spherocytosis and dehydrated hereditary spherocy- ability that causes the RBCs to be dehydrated.3 It is also known
tosis, and they can be divided into subtypes depending on as hereditary xerocytosis. It is the most common form of
whether or not the cation leak is temperature dependent.32 stomatocytosis.4
Pathophysiology. In DHSt, the RBC membrane is exces-
Overhydrated Hereditary Stomatocytosis. Overhy- sively permeable to potassium. The potassium leaks out of the
drated hereditary stomatocytosis (OHSt) is a rare hemolytic cell, but this is not balanced by an increase in sodium. Because
anemia due to a defect in membrane cation permeability that of the reduced intracellular cation concentration, water is lost
cause the RBCs to be overhydrated.3 It is inherited in an auto- from the cell.3,4 The specific defect has not been discovered, but
somal dominant pattern.10 the abnormal gene has been mapped in some patients to
Pathophysiology. In OHSt, the RBC membrane is exces- 16q23-24.4,32,34
sively permeable to sodium and potassium at 37 C.32 There Clinical and laboratory findings. Patients with DHSt
is an influx of sodium into the cell that exceeds the loss of generally have mild to moderate anemia, reticulocytosis, jaun-
potassium, which results in a net increase in the intracellular dice, and mild to moderate splenomegaly.4,10 Approximately
cation concentration. As a result, more water enters the cell, one third of patients have familial pseudohyperkalemia.4,10
and the cell swells and becomes stomatocytic. Because of the Fetal loss, hydrops fetalis, and neonatal hepatitis can also be
water influx, the cytoplasm has decreased density and viscosity. features of DHSt.4 The RBCs are dehydrated, as evidenced by
The increase in cell volume without an increase in membrane the elevated MCHC and decreased osmotic fragility.4,10 The
surface area causes premature hemolysis in the spleen.3 The RBC morphology includes stomatocytes (usually fewer than
exact molecular defect is unknown, but mutations in the RHAG 10%), target cells, burr cells, and RBCs in which the hemoglo-
gene that codes for RhAG protein, along with a deficiency of bin appears to be puddled in discrete areas on the cell periph-
the RhAG protein in the membrane, has been found in patients ery.4,10 Most patients with DHSt do not require treatment.
with OHSt.32,33 Most patients also have a deficiency of stoma- Splenectomy does not improve the anemia, and it is contrain-
tin, a transmembrane protein.32,33 Stomatin may participate in dicated because it increases the risk of thromboembolic
regulation of ion channels, but its role in OHSt is unclear.32 complications.3,4
The exact function of RhAG is unknown.
Clinical and laboratory findings. OHSt can cause Other Hereditary and Acquired Membrane Defects
moderate to severe hemolytic anemia. The diagnostic features with Stomatocytes
include 5% to 50% stomatocytes on the peripheral blood Rh Deficiency Syndrome. Rh deficiency syndrome
film (Figure 23-8), macrocytes (MCV of 110 to 150 fL), comprises a group of rare hereditary conditions in which the
324 PART IV Hematopathology: Erythrocyte Disorders

expression of Rh membrane proteins is absent (Rh-null) or anemia may resolve, however, if the patient is able to undergo
decreased (Rh-mod).4,13 Cases of the syndrome can be geneti- liver transplantation.4
cally divided into the amorph type (caused by mutations in Rh
proteins) and the regulatory type (caused by mutations in a Neuroacanthocytosis. Neuroacanthocytosis is a term
protein that regulates Rh gene expression).4,13 Patients with Rh used to describe a group of rare inherited disorders character-
deficiency syndrome present with mild to moderate hemolytic ized by neurologic impairment and acanthocytes on the
anemia. Stomatocytes and occasional spherocytes may be peripheral blood film. Three disorders are provided as exam-
observed on the peripheral blood film. Symptomatic patients ples in this group: abetalipoproteinemia, McLeod syndrome,
may be treated by splenectomy, which improves the anemia. and chorea acanthocytosis.35
Abetalipoproteinemia (ABL) is a rare autosomal recessive dis-
Acquired Stomatocytosis. Stomatocytosis occurs fre- order characterized by fat malabsorption, progressive ataxia,
quently as a drying artifact on Wright-stained peripheral blood neuropathy, retinitis pigmentosa, and acanthocytosis.35 ABL
films. A medical laboratory professional should examine many can first manifest with steatorrhea and failure to thrive in
areas on several films before categorizing the result as stomato- young children (due to fat malabsorption) or as ataxia and
cytosis, because in true stomatocytosis such cells should be neuropathy in young adults (due to decreased absorption of
found in all areas of the blood film. In normal individuals, 3% fat-soluble vitamin E).35ABL is caused by mutations in the MTP
to 5% of RBCs may be stomatocytes.4 In wet preparations in (microsomal triglyceride transfer protein) gene; MTP is needed
which RBCs are diluted in their own plasma and examined to transfer and assemble lipids onto apolipoprotein B.35 The
under phase microscopy, stomatocytes tend to be bowl shaped mutations result in an absence in the plasma of chylomicrons,
or uniconcave, rather than the normal biconcave shape. This very-low-density lipoproteins (which transport triglycerides),
technique can eliminate some of the artifactual stomatocytosis, and low-density lipoproteins (which transport cholesterol).13,35
but target cells also may appear bowl shaped in solution. Acute Consequently triglycerides and cholesterol are decreased, but
alcoholism and a wide variety of other conditions and medica- sphingomyelin is increased in the plasma.13 Because RBC
tions have been associated with acquired stomatocytosis.4 membrane lipids are in equilibrium with plasma lipids, the
RBC membrane acquires increased sphingomyelin, which
Other Hereditary and Acquired Membrane Defects decreases the fluidity of the RBC membrane and results in the
with Acanthocytes shape change. The shape defect is not present in developing
Acanthocytes (spur cells) are small, dense RBCs with a few nucleated RBCs or reticulocytes, but progresses as RBCs age in
irregular projections that vary in width, length, and surface the circulation.13 Other unknown mechanisms may also con-
distribution. These are distinct from burr cells (echinocytes), tribute to the formation of acanthocytes in ABL. Usually, 50%
which typically have small, uniform projections evenly distrib- to 90% of the RBCs are acanthocytes.4,13 Affected individuals
uted on the surface of the RBC (see Chapter 18). The differen- have a mild hemolytic anemia, normal RBC indices, and
tiation is easier to make on scanning electron micrographs and normal to slightly elevated reticulocyte counts.13 Early treat-
on wet preparations than on dried blood films.35,36 A small ment with high doses of vitamins A and E can reduce the
number of burr cells (less than 3% of RBCs) can be present on neuropathy and retinopathy.13 Patients with other related dis-
the peripheral blood films of healthy individuals, while orders, such as hypobetalipoproteinemia due to mutations in
increased numbers of burr cells are observed in uremia and the APOB gene, also may have acanthocytosis and neurologic
pyruvate kinase deficiency.13,35 Acanthocytes, however, are not disease.4,35
present on peripheral blood films of healthy individuals, but The McLeod syndrome (MLS) is an X-linked disorder caused
can be found in severe liver disease (spur cell anemia), neuro- by mutations in the KX gene.36 KX codes for the Kx protein, a
acanthocytosis (including abetalipoproteinemia), and other membrane precursor of the Kell blood group antigens.35 Men
conditions such as malnutrition and hypothyroidism.13 who lack Kx on their RBCs have reduced expression of Kell
antigens, reduced RBC deformability, and shortened RBC sur-
Spur Cell Anemia. A small percentage of patients with vival.35 Patients with MLS have variable acanthocytosis (up to
severe liver disease develop a hemolytic anemia with acantho- 85%), mild anemia, and late-onset (age 40 to 60 years), slowly
cytosis called spur cell anemia. The acanthocytes are due to a progressive chorea (movement disorders), peripheral neuropa-
defect in the lipid component of the membrane. In severe liver thy, myopathy, and neuropsychiatric manifestations.35,36 Some
disease, there is excess free cholesterol because of the presence female heterozygote carriers may have acanthocytes, but neuro
of abnormal plasma lipoproteins. An equilibrium is sought logic symptoms are rare. Clinical manifestations in females
between free cholesterol in the plasma and the RBC membrane depend on the proportion of RBCs with the normal X chromo-
cholesterol, and cholesterol preferentially accumulates in the some inactivated vs. those with the mutant X chromosome
outer bilayer leaflet of the membrane.4,13 The spleen remodels inactivated.13,36
the membrane into the acanthocyte shape.13 Acanthocytes have Chorea acanthocytosis (ChAc) is a rare autosomal recessive
long, rigid projections and become entrapped and hemolyzed disorder characterized by chorea, hyperkinesia, cognitive
in the spleen, which results in a rapidly progressive anemia of impairments, and neuropsychiatric symptoms.35 The mean age
moderate severity, splenomegaly, and jaundice.4,13 Spur cell of onset of the neurologic symptoms is 35 years.35 In patients
anemia in end-stage liver disease has a poor prognosis. The with ChAc, 5% to 50% of RBCs on the peripheral blood film
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 325

GPI-anchored
protein

Glycan core
Extracellular
domain

Phosphatidyl-
inositol

Lipid
bilayer

Intracellular
domain

Figure 23-9 Glycosylphosphatidylinositol (GPI) anchor for attachment of surface proteins to the cell membrane. Left, The structure of a GPI-anchored
protein. The GPI anchor consists of a phosphatidylinositol molecule in the outer leaflet of the lipid bilayer, which is connected to a glycan core consisting of
glucosamine, three mannose residues, and ethanolamine phosphate molecules. The protein is linked to the anchor at its C-terminus by an amide bond. The
result is a surface protein with a fluid and mobile attachment to the cell surface. The entire polypeptide is present in the extracellular milieu. Right, In contrast,
a transmembrane protein has an extracellular domain, a short transmembrane domain, and an intracellular domain. (From Ware RE, Rosse WF: Autoimmune
hemolytic anemia. In Nathan DG, Orkin SH, editors: Nathan and Oskis hematology of infancy and childhood, ed 5, Philadelphia, 1998, Saunders, p 514.)

are acanthocytes.35 ChAc is caused by mutations in VPS13A, a leaflet of the lipid bilayer. The glycan core consists of
gene that codes for chorein, a protein with uncertain cellular glucosamine, three mannose residues, and ethanolamine
functions.36 Chorein may be involved in trafficking proteins to phosphate molecules. Over 29 proteins are involved in the
the cell membrane; consequently a deficiency of chorein may biosynthesis of the GPI anchor.38 GPI-anchored proteins
lead to abnormal membrane protein structure and acanthocyte attach to the glycan core by an amide bond at their C-termini
formation.35 (Figure 23-9).
In PNH, a stem cell acquires a mutation in the PIGA
Acquired Membrane Defect: gene that codes for PI glycan class A (PIG-A).40 Over 180 dif-
Paroxysmal Nocturnal Hemoglobinuria ferent mutations have been identified in the PIGA gene in
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare chronic patients with PNH.38 The PIGA gene is located on the X chro-
intravascular hemolytic anemia caused by an acquired clonal mosome (Xp22.1) and codes for a glycosyltransferase needed
stem cell mutation that results in circulating blood cells that for the first step in the biosynthesis of the GPI anchor to add
lack glycosylphosphatidylinositol (GPI)anchored proteins, N-acetylglucosamine to PI.37,38 Without a fully functional
such as CD55 and CD59.37 The absence of CD55 and CD59 PIG-A glycosyltransferase, the cells are unable to synthesize the
on the surface of the RBCs renders them susceptible to spon- glycan core on the PI molecules effectively, and therefore the
taneous lysis by complement. Because the mutation occurs in cells are deficient in the surface GPI anchors. Without GPI
a stem cell, the defect is also found in platelets, granulocytes, anchors, all the progeny of the mutated stem cell are unable
monocytes, and lymphocytes.38 PNH is uncommon, with an to express any of the approximately 27 currently known GPI-
annual incidence of two to five new cases per million persons anchored proteins found on normal hematopoietic cells.38 The
in the United States.39 GPI-anchored proteins are accessory molecules for receptors,
ectoenzymes, adhesion molecules, or complement regula-
Pathophysiology of the Hemolytic Anemia tors.38 Relevant to the expression of the hemolysis in PNH, two
The GPI anchor consists of a phosphatidylinositol (PI) mole- GPI-anchored proteins are absent or deficient on the RBC
cule and a glycan core. The PI is incorporated in the outer membrane: decay-accelerating factor (DAF, or CD55) and
326 PART IV Hematopathology: Erythrocyte Disorders

membrane inhibitor of reactive lysis (MIRL, or CD59).39 CD55


and CD59 are complement-inhibiting proteins. CD55 inhibits BOX 23-2 Major Clinical Manifestations and
the formation and stability of the alternate pathway comple- Complications of Paroxysmal Nocturnal Hemoglobinuria
ment C3 and C5 convertases, and CD59 prevents the forma-
tion of the membrane attack complex. When CD55 and CD59 Related to Intravascular Hemolysis
are absent from the RBC surface, the cell is unable to prevent Anemia
the activation of complement, and spontaneous and chronic Hemoglobinuria
intravascular hemolysis occurs. Because the PIGA mutation is Fatigue
on the X chromosome, only one acquired mutation in a stem Chronic renal failure
cell is needed for the PNH phenotype (males have only one X Cholelithiasis
chromosome, and in females one of the X chromosomes is
inactivated).37 Related to Thrombophilia
In most patients, the PIGA mutant clone coexists with Venous thrombosis
normal stem cells and progenitors, which results in a popula- Abdominal vein thrombosis: hepatic (Budd-Chiari syndrome),
tion of RBCs that are GPI deficient and a population that is splenic, renal veins
normal. Some patients have a greater expansion of the mutant Portal hypertension
clone, a higher percentage of circulating GPI-deficient RBCs, Cerebral vein thrombosis
and a more severe chronic hemolytic anemia.37 On the other Retinal vein thrombosis and loss of vision
hand, other patients have minimal expansion of the mutant Deep vein thrombosis, pulmonary emboli
clone and have a lower percentage of circulating GPI-deficient Arterial thrombosis (less common)
RBCs. These patients may be asymptomatic and may not Stroke
require any treatment.37 Myocardial infarction
Patients with PNH also display phenotypic mosaicism.37
The mosaicism results when a single patient is able to harbor Related to Bone Marrow Failure
normal clones as well as mutant clones with different PIGA Pancytopenia: fatigue, infections, bleeding
mutations, and those different mutations result in variable Myelodysplastic syndrome
expression of CD55 and CD59 on the RBCs. The RBC pheno- Acute myeloid leukemia (rare)
types in PNH can be classified as type I, type II, or type III.37,38 Adapted from Bessler M, Hiken J: The pathophysiology of disease in patients with
Type I RBCs are phenotypically normal, express normal paroxysmal nocturnal hemoglobinuria, Hematology Am Soc Hematol Educ
amounts of CD55 and CD59, and undergo little or no Program, pp 104-110, 2008.
complement-mediated hemolysis. Type II RBCs are the result
of a PIGA mutation that causes only a partial deficiency of
CD55 and CD59, and these cells are relatively resistant to Laboratory Findings
complement-mediated hemolysis. Type III RBCs are the result Biochemical evidence of intravascular hemolysis includes
of a PIGA mutation that causes a complete deficiency of GPI, hemoglobinemia, hemoglobinuria, decreased level of serum
and therefore no CD55 and CD59 proteins are anchored to haptoglobin, increased levels of serum indirect bilirubin and
the RBC surface. Type III RBCs are highly sensitive to spontane- lactate dehydrogenase, and hemosiderinuria (see Chapter 22).
ous lysis by complement. Therefore, when the severity of Hemoglobinuria is present in only 25% of patients at diagno-
the hemolysis in PNH is being assessed, both the relative sis, but will occur in most patients during the course of the
amount and the type of circulating RBCs are important illness.41 Only a small minority of patients report episodic
considerations.37 hemoglobinuria during the night.39 Hemosiderinuria is an
In addition to hemolysis, patients with PNH may have bone indication of chronic intravascular hemolysis and may be
marrow dysfunction that contributes to the severity of the detected with Prussian blue staining of the urine sediment.
anemia. Many patients have a history of bone marrow failure Reticulocyte counts are mildly to moderately increased,
caused by acquired aplastic anemia or myelodysplastic syn- with less elevation than would be expected in other hemolytic
drome that precedes or coincides with the onset of PNH.41 anemias of comparable severity. The MCV may be slightly
elevated, which reflects reticulocytosis. Osmotic fragility is
Clinical Manifestations normal, and the DAT result is negative. If the patient does not
The onset of PNH most frequently occurs in young adulthood, receive transfusions, iron deficiency develops due to the loss
but can occur in childhood and advanced age.38 The major of hemoglobin iron in the urine, and the RBCs become micro-
clinical manifestations and complications of PNH are those cytic and hypochromic. Serum iron studies (serum iron, total
associated with hemolytic anemia, thrombophilia, and bone iron-binding capacity, and serum ferritin) are performed to
marrow failure (Box 23-2).38 Anemia is mild to severe depend- detect iron deficiency (see Chapter 19). Folate deficiency
ing on the degree of hemolysis and the presence of bone often occurs if there is chronic erythroid hyperplasia and a
marrow failure. Hepatic vein thrombosis (Budd-Chiari syn- greater need for folate, which leads to secondary macrocytosis.
drome) is the most common manifestation of thrombophilia Pancytopenia may occur if there is concomitant bone marrow
and is a serious, often fatal complication.39 failure.
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 327
The bone marrow aspirate and biopsy specimen are
examined for abnormal cells, evidence of an underlying bone
marrow failure syndrome, and cytogenetic abnormalities.41
The bone marrow may be normocellular to hypercellular with
erythroid hyperplasia in response to the hemolysis, or it may
be hypocellular in concomitant bone marrow failure.39 A
finding of dysplasia or certain chromosome abnormalities is
helpful in diagnosis of a myelodysplastic syndrome (see
Chapter 35). An abnormal karyotype is found in 20% of
patients with PNH.39
Confirmation of PNH requires demonstration of GPI-
deficient cells in peripheral blood RBCs and granulocytes.38
Flow cytometric analysis of RBCs with fluorescence-labeled
anti-CD59 or anti-CD55 determines the proportion of types I,
II, and III RBCs, and thus can provide an assessment of the
severity of the hemolysis (Figure 23-10).37,42 Type I cells with
normal expression of CD59 show the highest intensity level of
fluorescence; type II cells with a partial deficiency of CD59
show moderate fluorescence; and type III cells with no CD59
are negative for fluorescence. Patients with a greater proportion
of type III cells (complete deficiency of GPI-anchored proteins)
are expected to have a high-grade hemolysis.37,41 Patients with
a high percentage of type II cells (partial deficiency of GPI-
anchored proteins) and a low percentage of type III cells may
have only modest hemolysis.37,41 The drawback of this method,
however, is decreased sensitivity and underestimation of the
number of type III cells, because RBCs lacking CD59 and CD55
undergo rapid intravascular lysis.41
Identification by flow cytometry of a deficiency of at least
two GPI-anchored proteins on granulocytes, such as CD59,
CD55, CD24, CD15, is a sensitive and reproducible method
for diagnosis of PNH.38 An alternative flow cytometric method
uses a fluorescein-labeled proaerolysin variant (FLAER).38
FLAER binds to the glycan core of the GPI anchor with a high
signal-to-noise ratio. The absence of binding on granulocytes
is indicative of GPI deficiency.38,41 Combining the FLAER assay
with multiparameter flow cytometry for CD45, CD33, and
CD14 increases sensitivity and specificity for detection of GPI-
deficient granulocytes and monocytes.42,43 Figure 23-10 Immunophenotypic analysis of erythrocytes in paroxysmal
The sugar water test (sucrose hemolysis test) and the Ham nocturnal hemoglobinuria by flow cytometry with fluorescence-labeled anti-
test (acidified serum lysis test) have insufficient sensitivity for CD59. Examples of surface CD59 expression are shown for three patients
diagnosis of PNH and have been replaced by flow cytometric and a control subject. The x-axis represents fluorescence intensity and the
techniques. y-axis represents the relative number of cells. A, Patient with type I and type
III erythrocytes. B, Patient with type I, II, and III erythrocytes. C, Patient with
Classification predominantly type III erythrocytes and a few type II cells. D, Type I cells in
The International PNH Interest Group proposed three subcat- a healthy control. Type I cells have normal expression of CD59 and show
the highest intensity level of fluorescence. (Adapted from Ware RE, Rosse
egories of PNH: (1) classic PNH, (2) PNH in the setting of
WF, Hall SE: Immunophenotypic analysis of reticulocytes in paroxysmal
another specified bone marrow disorder, and (3) subclinical
nocturnal hemoglobinuria, Blood 86:1586, 1995.)
PNH.41 In classic PNH, there is clinical and biochemical evi-
dence of intravascular hemolysis, reticulocytosis, a cellular
bone marrow with erythroid hyperplasia and normal morphol- usually less than 30% of the total neutrophils. In subclinical
ogy, and a normal karyotype. In addition, more than 50% of PNH, patients have no clinical or biochemical evidence of
the circulating neutrophils are GPI-deficient.41 In PNH in the hemolysis but have a small subpopulation of GPI-deficient
setting of another bone marrow disorder, patients have evi- neutrophils that comprise less than 1% of the total circulating
dence of hemolysis, but have a history of or concomitant aplas- neutrophils.41 This subcategory is found in association with
tic anemia, myeloproliferative disorder, or other myelopathy. bone marrow failure syndromes. Features of each subgroup are
The number of GPI-deficient neutrophils are variable, but are summarized in Table 23-3.
328 PART IV Hematopathology: Erythrocyte Disorders

TABLE 23-3 Classification of Paroxysmal Nocturnal Hemoglobinuria (PNH)*


% GPI-APDeficient
Subcategory Rate of Intravascular Hemolysis Bone Marrow Neutrophils
Classic PNH Marked; visible hemoglobinuria frequent Normocellular to hypercellular with >50%
erythroid hyperplasia and normal
or near-normal morphology
PNH in the setting of another Mild to moderate; visible hemoglobinuria Evidence of a concomitant bone Variable; usually <30%
specified bone marrow disorder is intermittent or absent marrow failure syndrome
Subclinical PNH No clinical or biochemical evidence of Evidence of a concomitant bone <1%
intravascular hemolysis marrow failure syndrome

Adapted from Parker CJ: Paroxysmal nocturnal hemoglobinuria. In Kaushansky K, Lichtman MA, Beutler TJ, etal, editors: Williams hematology, ed 8, New York, 2010, McGraw-
Hill Medical, chap 40. Available at: http://www.accessmedicine.com/content.aspx?aID=6109660. Accessed August 18, 2010.
GPI-AP, Glycosylphosphatidylinositol-anchored protein.
*Subclassification proposed by the International PNH Interest Group (Parker C, Omine M, Richards S, etal: Diagnosis and management of paroxysmal nocturnal hemoglobinuria,
Blood 106:3699-3709, 2005).

Bone marrow failure syndromes include aplastic anemia, refractory anemia/myelodysplastic syndrome, other myelopathy (e.g., myelofibrosis).

Determined by very sensitive flow cytometric analysis.

Treatment
RED BLOOD CELL ENZYMOPATHIES
In 2007, the U.S. Food and Drug Administration approved
eculizumab for the treatment of hemolysis in PNH.44 Eculi- The major function of the RBCs is to transport oxygen to the
zumab is a humanized monoclonal antibody that binds to tissues over their life span of 120 days. For the RBCs to do that
complement C5, prevents its cleavage to C5a and C5b, and effectively, they need functional enzymes to maintain glycoly-
thus inhibits the formation of the membrane attack unit.37 sis, preserve the shape and deformability of the cell membrane,
Eculizumab is the treatment of choice for patients with classic keep hemoglobin iron in a reduced state, protect hemoglobin
PNH. It results in an improvement of the anemia and a decrease and other cellular proteins from oxidative denaturation, and
in transfusion requirements.44 There is also a reduction in the degrade and salvage nucleotides. Deficiencies in RBC enzymes
lactate dehydrogenase level in the serum.44 Patients taking ecu- may impair these functions to varying degrees and decrease the
lizumab have an increased risk for infections with Neisseria life span of the cell. The most important metabolic pathways
meningitidis and need to be vaccinated prior to administration are the Embden-Meyerhof pathway (anaerobic glycolysis) and
of the drug.44 Mild to moderate anemia and reticulocytosis the hexose monophosphate (pentose) shunt45 (see Figure 9-1).
persist in some patients, likely due to C3 opsonization and The most commonly encountered enzymopathies are deficien-
extravascular hemolysis, because eculizumab does not inhibit cies of glucose-6-phosphate dehydrogenase and pyruvate
complement C3.37 Eculizumab is not curative and does not kinase. Other RBC enzymopathies are rare.45
address the thrombophilic and bone marrow failure complica-
tions of PNH. Glucose-6-Phosphate Dehydrogenase
Other treatments for PNH are mainly supportive and Deficiency
include transfusions, antibiotics, and anticoagulants. Iron A major function of glucose-6-phosphate dehydrogenase
therapy is given to help alleviate the iron deficiency caused (G6PD) is to keep hemoglobin iron in the reduced, physiologi-
by the urinary loss of hemoglobin, and folate supplementation cally active (ferrous) state for oxygen transport and to protect
is given to replace the folate consumed in accelerated erythro- hemoglobin from oxidative damage. In the hexose monophos-
poiesis. Administration of androgens and glucocorticoids to phate shunt, G6PD generates reduced nicotinamide adenine
ameliorate anemia is controversial.37 Anticoagulants are used dinucleotide phosphate (NADPH) while oxidizing glucose-6-
in the treatment of thrombotic complications. In suitable phosphate (Figure 23-11). The NADPH restores reduced gluta-
patients, hematopoietic stem cell transplantation with an HLA- thione (GSH) from oxidized glutathione (GSSG) through the
matched sibling donor may be an option and can be a curative transfer of a hydrogen ion by glutathione reductase. GSH is
therapy, with an overall survival of 50% to 60% in such crucial in reducing hydrogen peroxide and oxygen radicals to
patients.37 maintain hemoglobin iron in the reduced (ferrous) state and
PNH is a serious disease, and most patients eventually die protect it from oxidative denaturation.45,46 G6PD provides the
from its various complications, such as thrombotic episodes or only means of generating the reduced form of nicotinamide,
pancytopenia. Thrombotic complications are the major cause NADPH, and in its absence the erythrocyte is particularly vul-
of morbidity and mortality in PNH.37 The incidence of trans- nerable to oxidative damage. Most oxidant drugs enable inter-
formation to myelodysplastic syndrome or acute leukemia is action between molecular oxygen and components of the RBC.
5% and less than 1%, respectively.37 The median survival after RBCs with normal G6PD activity are able to detoxify the oxida-
diagnosis is approximately 10 years.37 tive compounds and safeguard the hemoglobin.
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 329

Oxidant H2O2 H2O isoenzymes have been identified.47 The normal G6PD variant
Glutathione peroxidase is designated G6PD-B. Some other G6PD variants are not asso-
ciated with significantly reduced enzyme activity in the RBCs.
Reduced glutathione (GSH) Oxidized glutathione (GSSG) A common mutant form that produces no clinical effects, leads
to no enzyme deficiency, and is present in 20% to 40% of
Glutathione reductase
individuals of African descent is G6PD-A+. The different vari-
ants of G6PD have been divided into classes by the World
NADP NADPH Health Organization, based on clinical symptoms and amount
of enzyme activity.52
Glucose-6-phosphate 6-phosphogluconate Class I includes rare variants with severe enzyme deficiency
Glucose-6-phosphate
dehydrogenase that give rise to chronic nonspherocytic hemolytic anemia
Figure 23-11 Function of glucose-6-phosphate dehydrogenase in (CNSHA).
generating reduced glutathione. Glucose-6-phosphate from the Embden- Class II includes variants with severe deficiency (less than
Meyerhof pathway is converted to 6-phosphogluconate by the action of 10% enzyme activity) and no CNSHA; they can lead to
glucose-6-phosphate dehydrogenase. In the reaction, oxidized NADP is severe, episodic acute hemolytic anemia associated with
reduced to NADPH. NADPH is then available to reduce oxidized glutathione infections and certain drugs and foods. G6PD-Mediterranean
(GSSG) to reduced glutathione (GSH) by the action of glutathione reductase. is a variant in this class.
Reduced glutathione is then available to reduce peroxide to water by the Class III includes variants with moderate deficiency (10% to
action of glutathione peroxidase. 60% enzyme activity) and no CNSHA; they can lead to
episodic acute hemolytic anemia associated with infections
The G6PD gene is located on the X chromosome and shows and certain drugs, but it is usually self-limited. G6PD-A
a characteristic X-linked pattern. Men can be normal hemizy- (found in approximately 10% of African American men)
gotes (have the normal allele) or deficient hemizygotes (have and G6PD-Canton (prevalent in Southeast Asia) are vari-
a variant allele). Women can be normal homozygotes (both ants in this class.
alleles normal), deficient homozygotes (both alleles abnor- Class IV includes variants with normal enzyme activity
mal), or heterozygotes (one normal allele and one abnormal (60% to 150%) associated with no hemolysis. G6PD-B and
allele).47 The level of G6PD enzyme in female heterozygotes G6PD-A+ are variants in this class.
lies between normal and deficient due to the random inactiva- Class V includes variants with increased enzyme activity
tion of one of the X chromosomes in each cell (lyonization). (greater than 150%), but this category is not clinically
Therefore the RBCs of female heterozygotes are a mosaic, with relevant.
some cells having normal G6PD activity and some cells having
deficient G6PD activity. Because the X inactivation is random, Pathophysiology
the proportions of normal and G6PD-deficient RBCs vary G6PD-deficient RBCs cannot generate sufficient NADPH to
among different women. Some heterozygous women may have reduce glutathione, and thus cannot effectively detoxify the
hemolytic attacks if they have a high proportion of G6PD- hydrogen peroxide produced upon exposure to oxidative stress.
deficient RBCs. Although this disorder is X-linked recessive Consequently oxidation of membrane thiols and hemoglobin
genetically, because heterozygous women have a decreased occurs. Hemoglobin is first oxidized to methemoglobin and
amount of the enzyme, it is biochemically codominant.47 then to Heinz bodies, intracellular precipitates of denatured
G6PD deficiency is the most common RBC enzyme defect hemoglobin that adhere to the inner RBC membrane. The
with a prevalence of 5% of the global population or approxi- mechanism of Heinz body formation is complex and is not
mately 329 million people worldwide.48 The prevalence in completely understood. Because of the membrane damage and
Africa, the Middle East, and the Americas is 7.5%, 6.0%, and loss of deformability, RBCs with Heinz bodies are rapidly
3.4%, respectively.48 G6PD deficiency has the highest preva- removed from the circulation by intravascular and extravascular
lence in geographic areas in which malaria is endemic because hemolysis.47 Reticulocytes have approximately five times
of the selective pressure of malaria.49 Studies in Africa show more G6PD activity than older RBCs, because enzyme activity
that G6PD deficiency (A variant) in hemizygous males confers decreases as the cells age. Therefore, during exposure to oxidants,
protection against life-threatening P. falciparum malaria.50 This the older RBCs with less G6PD are preferentially hemolyzed.49
protective effect is not observed in heterozygous females
because of their mosaicism of normal and G6PD-deficient Clinical Manifestations
RBCs.50 In a recent case-control study in South Asia, G6PD The vast majority of individuals with G6PD deficiency are
deficiency (Mediterranean variant) conferred protection against asymptomatic throughout their lives. However, some patients
Plasmodium vivax infection, with a greater protective effect in have clinical manifestations. The following three clinical syn-
hemizygous males.51 dromes are recognized: (1) acute hemolytic anemia, (2) neo-
There are over 140 known mutations in the G6PD gene, and natal jaundice (hyperbilirubinemia), and (3) CNSHA.49
most are point mutations.46Amino acid substitutions change
the primary protein structure of the enzyme and thus affect its Acute Hemolytic Anemia. Oxidative stress can precipi-
function, stability, or both. In addition, more than 400 variant tate a hemolytic episode, and there are three main triggers:
330 PART IV Hematopathology: Erythrocyte Disorders

sulfhydryl groups. Infectious agents implicated in hemolytic


BOX 23-3 Drugs with Strong Evidence-Based episodes include bacteria, viruses, and rickettsia.
Support for an Association with Drug-Induced Hemolysis Favism is a severe hemolytic episode that occurs in a small
in Glucose-6-Phosphate Dehydrogenase Deficiency percentage of G6PD-deficient individuals after exposure to the
Dapsone fava bean. Favism can initially manifest with a sudden onset
Methylthioninium chloride (methylene blue) of acute intravascular hemolysis within hours of exposure to
Nitrofurantoin the fava bean or with a gradual onset within 24 to 48 hours of
Phenazopyridine exposure.47 Hemoglobinuria is one of the first signs of the
Primaquine disorder. Only a small percentage of G6PD-deficient individu-
Rasburicase als manifest favism, and most of these have the G6PD-
Tolonium chloride (toluidine blue) Mediterranean variant.
From Youngster I, Arcavi L, Schechmaster R, etal: Medications and glucose-6-
phosphate dehydrogenase deficiency: an evidence-based review, Drug Saf Neonatal Hyperbilirubinemia. Neonatal hyperbilirubine-
33:713-726, 2010. mia secondary to G6PD deficiency occurs particularly in Greece,
Sardinia, Africa, and the United States. Anemia is usually not
present at birth, and jaundice generally appears 2 to 3 days
(1) certain drugs, (2) infections, and (3) fava beans. Hemolysis after birth.49 These infants must be closely monitored, because
secondary to drug exposure is the classic manifestation of the hyperbilirubinemia can be severe and cause bilirubin
G6PD deficiency. The actual discovery of G6PD deficiency was encephalopathy (kernicterus).47 Severe hyperbilirubinemia
a direct consequence of investigations into the development of occurs more often in infants who, in addition to a mutated
hemolysis after ingestion of the antimalarial agent primaquine G6PD gene, are homozygous for a mutation in the promoter
by some individuals. Box 23-3 lists drugs for which there is region of the bilirubin-uridine diphosphoglucuronate glucuro-
strong evidence-based support for an association with drug- nosyltransferase 1 (UGT1A1) gene, which impairs their ability
induced hemolysis in G6PD-deficient individuals.53 With some to conjugate and excrete indirect bilirubin.47 In addition,
of these drugs, hemolysis occurs in some individuals and not genetic factors, which include the particular variant of G6PD,
in others, and different dosages give rise to different degrees of and environmental factors (e.g., drugs given to the mother or
hemolysis, depending on other circumstances, such as a infant, the presence of infection, and gestational age) probably
coexisting infection or concomitant use of other drugs.53 Also, play an important role in the occurrence of neonatal hyperbili-
because reticulocytes have higher amounts of G6PD activity rubinemia, because a wide variation exists in frequency and
and are more resistant to oxidative stress, individuals with high severity in different populations with G6PD deficiency.
reticulocyte counts have a lower rate of hemolysis.53
Individuals with class II and III G6PD deficiency are clini- Chronic Nonspherocytic Hemolytic Anemia. Most
cally and hematologically normal until the offending drug is patients with G6PD deficiency have no clinical symptoms, and
taken. Clinical hemolysis can begin abruptly within hours or their RBCs have only a slightly reduced life span. A small per-
occur gradually 1 to 3 days after the drug is taken.47,49 A rapid centage of G6PD-deficient patients have CNSHA, as evidenced
drop in hemoglobin may occur, and the anemia can range by chronic hyperbilirubinemia, decreased serum haptoglobin
from mild to very severe. Hemoglobinuria is a chief symptom level, and increased lactate dehydrogenase level. Most of these
and indicates that the hemolysis is intravascular, although patients are diagnosed at birth as having neonatal hyperbiliru-
some extravascular hemolysis probably also takes place. Heinz binemia, and the hemolysis continues into adulthood. They
bodies can be observed by incubating the blood in a supravital usually do not have hemoglobinuria, which suggests that the
stain. Reticulocytes increase within 4 to 6 days.47 Generally, chronic hemolysis is extravascular as opposed to intravascular.
with class III variants such as G6PD-A, the hemolytic episode There is evidence that normal oxidative stress causes the sulf-
is self-limiting because the newly formed reticulocytes have hydryl groups of the membrane proteins (especially spectrin)
higher G6PD activity. With class II variants such as G6PD- to be oxidized and conformational changes to occur so that
Mediterranean, the RBCs are severely G6PD-deficient, so the the RBCs are removed by the spleen, probably by the same
hemolytic episode may be longer and the hemolysis is not mechanism as in HS. The RBC morphology is unremarkable
self-limiting.47 and is referred to as nonspherocytic. These patients also are vul-
Infection is probably the most common cause of hemolysis nerable to acute oxidative stress from the same agents as those
in individuals with G6PD deficiency. During the episode, the affecting other G6PD patients and may have acute episodes of
hemoglobin can drop 3 to 4g/dL if reticulocyte production is hemoglobinuria. The severity of CNSHA is extremely variable,
suppressed by the infection. The hemolysis resolves when probably because of the different mutations that can occur in
infection clears.47 The mechanism of hemolysis induced by the G6PD gene.
acute and subacute infection is poorly understood, but the
generation of hydrogen peroxide by phagocytizing leukocytes Laboratory Findings
may play a role.47 Diminished liver function may contribute The anemia occurring during a hemolytic crisis may range
further to the oxidant stress on the RBCs by allowing the accu- from moderate to extremely severe and is usually normocytic,
mulation of metabolites capable of oxidizing RBC membrane normochromic. The morphology of G6PD-deficient RBCs is
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 331

Figure 23-12 Principle of the glucose-6-phosphate dehydrogenase (G6PD) deficiency test. In the screening test, reduced nicotinamide adenine dinucleotide
phosphate (NADPH) fluoresces, whereas the unreduced form (NADP) does not. If glucose-6-phosphate is oxidized to 6-phosphogluconate, the coenzyme NADP
is reduced to NADPH, and a corresponding increase in fluorescence occurs.

normal except during a hemolytic episode. The change in mor- Treatment and Prognosis
phology during a hemolytic episode varies depending on the Therapy for patients with G6PD deficiency involves preventing
amount of hemolysis. In some patients, the change is not strik- the common manifestations of hemolytic anemia and neona-
ing, but in other individuals with severe variants, marked tal jaundice. Most hemolytic episodes, especially in individuals
anisocytosis, poikilocytosis, spherocytosis, and schistocytosis with G6PD-A, are self-limited. In patients with more severe
may occur.47 Bite cells (RBCs in which the margin appears types, such as G6PD-Mediterranean, this is not always the case.
indented, purportedly due to splenic removal of a Heinz body) Screening is important in populations that have a high inci-
may be observed in rare cases in some patients with drug- dence of this deficiency. Because neonatal hyperbilirubinemia
induced hemolysis, but should not be considered a specific cannot be prevented, it must be sought in the populations that
feature of G6PD deficiency.47,54 Bite cells are absent in acute are at high risk and must be treated immediately. Neonatal
and chronic hemolytic states associated with common G6PD- hyperbilirubinemia is not a major problem in most popula-
deficient variants, and they can also be found in other condi- tions in the United States. The prevention of acute hemolytic
tions.47,54 Heinz bodies cannot be detected with Wright staining, anemia is difficult, because multiple causes exist; however,
but when exposed to certain basic dyes or supravital stains, some cases of acute hemolytic anemia are easily preventable,
such as crystal violet, they assume a purple color and appear such as by avoidance of fava bean consumption in families in
at the margin of the RBC (see Figure 14-11). The reticulocyte which this sensitivity exists.
count is increased and may reach 30% of RBCs. Consistent Favism is a relatively dangerous disease, and fatalities were
with intravascular hemolysis, the serum haptoglobin level is common before transfusion services were available. Prevention
severely decreased, and there is hemoglobinemia and hemo- of drug-induced disease is possible by choosing alternate drugs
globinuria. The unconjugated (indirect) bilirubin level is also when possible. In cases in which the offending drugs must be
elevated. The white blood cell (WBC) count is moderately used, especially in individuals with G6PD-A, the dosage can
elevated, and the platelet count varies. be lowered to decrease the hemolysis to a manageable level.
Quantitative assays of G6PD enzyme activity in RBCs can Infection-induced hemolysis is more difficult to prevent but
be performed to determine the severity of the G6PD deficiency, can be detected early in the course of the episode and treated
but screening tests are usually adequate. Both tests are based if necessary. Most episodes resolve without treatment but may
on the reduction of an oxidized pyridine nucleotide (NADP to be severe enough to warrant a blood transfusion. In patients
NADPH) during the reaction shown in Figure 23-12. In the with hemoglobin levels greater than 9 g/dL with persistent
quantitative assay, hemolysate of the patients blood is added hemoglobinuria, close monitoring is important. Neonates with
to a mixture of reagents that has been constituted in such a severe hyperbilirubinemia and jaundice secondary to G6PD
way that the amount of enzyme represents the limiting step; deficiency may require exchange transfusion.
the activity is measured by the change in absorbance at 340nm.
The principle of the screening test is the same except that, Differential Diagnosis
rather than measuring the absorbance by the reduced NADPH, The drug-induced hemolysis associated with G6PD deficiency
visual observation of the fluorescence of reduced nucleotide involving class II and III variants must be differentiated from
when activated with ultraviolet light is used to evaluate whether other drug-induced hemolytic anemias. This differentiation
pyridine nucleotide (NADP) has been reduced. This is done can be accomplished by means of the screening test or quan-
by mixing the blood and reagents, placing them on a filter titative assay for G6PD activity. Patients with CNSHA (class I
paper, and observing the filter paper under ultraviolet light. variant G6PD deficiency) must be differentiated from patients
Because reticulocytes have higher G6PD levels than mature with HS, hemoglobinopathies, and other enzymopathies.
RBCs, assays or screening tests should not be performed on
samples collected after an individual has had a severe hemo- Pyruvate Kinase Deficiency
lytic crisis, because the G6PD levels may be falsely elevated due Pyruvate kinase (PK) is a rate-limiting key enzyme of the gly-
to reticulocytosis. The testing should be performed after the colytic pathway of RBCs. It catalyzes the conversion of phos-
reticulocyte and total RBC counts have returned to normal. phoenolpyruvate to pyruvate, forming ATP (see Figure 9-1). PK
Patients who are not G6PD deficient are expected to have high deficiency is an uncommon disorder with an estimated preva-
G6PD activity during episodes of reticulocytosis. Therefore a lence of 1 per 200,000 in the white population47,55; however,
normal rather than an increased result on a G6PD assay in a of the hereditary chronic nonspherocytic hemolytic anemias,
patient with reticulocytosis provides a clue that the patient may it occurs with the most frequency.47 PK deficiency is an auto-
be G6PD deficient. somal recessive disorder, and symptomatic hemolytic anemia
332 PART IV Hematopathology: Erythrocyte Disorders

occurs in homozygotes or compound heterozygotes. PK defi- platelet counts are normal or slightly increased. Patients usually
ciency occurs in high frequency in five Amish kindreds in have the characteristic laboratory findings of chronic hemoly-
Pennsylvania; all affected family members are homozygous for sis, including an increased serum indirect bilirubin level, a
the same mutation.56 Over 150 mutations (mainly missense) decreased serum haptoglobin level, and increased urinary uro-
have been reported in PKLR, the gene that codes for PK.57 bilinogen. The osmotic fragility of fresh cells is usually normal,
and the DAT result is negative.
Pathophysiology The diagnosis of PK deficiency depends on the specific
The exact mechanisms for hemolysis and decreased RBC life demonstration of quantitatively reduced activity or qualitative
span in PK-deficient cells are poorly understood. The metabolic abnormalities of erythrocyte PK. The enzyme can be measured
consequences of PK deficiency are ATP depletion and an by spectrophotometric assay of a hemolysate prepared from
increase in 2,3-bisphosphoglycerate (2,3-BPG).58 The increase RBCs after careful removal of WBCs. WBCs have a very high
in 2,3-BPG shifts the hemoglobin-oxygen dissociation curve to PK level, and contamination of the hemolysate with WBCs
the right and decreases the oxygen affinity of hemoglobin58 falsely increases the result (i.e., in a PK deficiency, the result
(see Chapter 10). This promotes greater release of oxygen to could be falsely normal).57 The assay uses phosphoenolpyru-
the tissues and enables affected individuals to tolerate lower vate, adenosine diphosphate (ADP), lactate dehydrogenase,
levels of hemoglobin.57,58 and the reduced form of nicotinamide adenine dinucleotide
(NADH), constituted in such a way that the oxidation of
Clinical Manifestations NADH is followed by measuring the change in absorbance at
Individuals with PK deficiency have a wide range of clinical 340nm and absorbance is decreased according to the amount
presentations, varying from severe neonatal anemia requiring of NADH oxidized to nicotinamide adenine dinucleotide
exchange or multiple transfusions to a fully compensated (NAD). More complex techniques may be necessary when a
hemolytic process in apparently healthy adults.58 Most patients, variant form of PK is suspected.57 Screening tests for PK defi-
however, have manifestations of chronic hemolysis, including ciency are based on the same principle as that described earlier
anemia, jaundice, splenomegaly, and increased incidence of except that the hemolysate and reagents are absorbed onto
gallstones (due to the production of excessive bilirubin).45 filter paper, and the loss of fluorescence is visually evaluated to
Rarely, folate deficiency (due to accelerated erythropoiesis), determine the oxidation of NADH (Figure 23-13).60 Mutation
bone marrow aplasia (usually due to parvovirus B19 infec- detection can be accomplished by sequencing the exons, flank-
tion), and skin ulcers can occur.54,57 Iron overload can occur ing regions, and promoter region.57 Molecular diagnosis is
even in the absence of transfusions.57,59 superior in sensitivity and specificity, is applicable for use in
prenatal testing, and enables correlation of certain mutations
Laboratory Findings with disease severity.57
The hemoglobin level is variable depending on the extent of
the hemolysis. Reticulocytosis is usually present, but not in Treatment and Prognosis
proportion to the severity of the anemia, because the reticulo- No specific therapy is available for PK deficiency except sup-
cytes are preferentially sequestered in the spleen.57 After sple- portive treatment and RBC transfusion as necessary. Splenec-
nectomy, there is an increase in the number of circulating tomy is beneficial in severe cases, and after this procedure the
reticulocytes, with a median count of approximately 800 hemoglobin level usually increases enough to reduce or elimi-
109/L.57 In addition to showing anisocytosis, poikilocytosis, nate the need for transfusion.54
and polychromasia, the peripheral blood film reveals a variable
number of burr cells, or echinocytes (in the range of 3% to Differential Diagnosis
30%), which increase in number after splenectomy.57 The post- Because PK deficiency causes chronic hemolytic anemia, it
splenectomy peripheral blood film may also show Howell-Jolly must be differentiated from hemoglobinopathies, HS, and
bodies, Pappenheimer bodies, and target cells. The WBC and other enzymopathies. An appropriate diagnostic strategy is first

Figure 23-13 Principle of the pyruvate kinase (PK) test. In the screening test, a red blood cell (RBC) suspension, made from blood with the plasma and
buffy coat removed, is incubated with a reagent that contains adenosine diphosphate (ADP), phosphoenolpyruvic acid, the reduced form of nicotinamide
adenine dinucleotide (NADH), and lactate dehydrogenase. When PK is present, the NADH is oxidized to NAD, which results in a loss of fluorescence when
spots made on filter paper are viewed under ultraviolet light. When pyruvate is deficient in the sample, the NADH remains reduced, and no loss of fluorescence
occurs.
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 333
to eliminate the hemoglobinopathies and HS from consider- aldolase, triosephosphate isomerase, phosphoglycerate kinase,
ation and then to proceed to tests for enzyme disorders. and enolase. All of these deficiencies are autosomal recessive
conditions except for phosphoglycerate kinase deficiency,
Other Enzymopathies which is X-linked, and enolase, which is presumably autosomal
Other RBC enzymopathies are rarely encountered. In addition dominant. Lactate dehydrogenase deficiency is not associated
to PK deficiency, deficiencies of other enzymes of the RBC with hemolysis. Deficiencies of 2,3-BPG mutase and 2,3-BPG
Embden-Meyerhof pathway have been described, including phosphatase are associated with mild erythrocytosis secondary
hexokinase, glucose phosphate isomerase, phosphofructokinase, to the near-absence of 2,3-BPG.

SUMMARY
The RBC membrane must have deformability for the RBC to overhydrated with decreased cytoplasmic viscosity, and stomato-
maneuver through the microcirculation and the splenic sieve over cytes are observed on the peripheral blood film. In DHSt, the RBCs
its life span of 120 days. The cellular properties that enable are dehydrated with increased cytoplasmic viscosity, and the
deformability are the biconcave, discoid shape of the cell; the peripheral blood film shows burr cells, target cells, few stomato-
viscoelasticity of the membrane; and cytoplasmic viscosity. cytes, and cells with puddled hemoglobin at the periphery. Sto-
Two transmembrane protein complexes, the ankyrin complex and matocytosis may also occur in Rh deficiency syndrome and in a
protein 4.1 junctional complex, provide vertical structural integrity variety of acquired conditions.
to the cell by anchoring the lipid bilayer to the underlying spectrin Acquired acanthocytosis can occur in severe liver disease (spur
skeleton. -Spectrin, -spectrin, and their accessory proteins cell anemia). Neuroacanthocytosis comprises a group of inherited
form a two-dimensional lattice to provide horizontal mechanical disorders characterized by neurologic impairment and the pres-
stability to the membrane. ence of acanthocytes on the peripheral blood film. Major disorders
HS is caused by mutations that disrupt the vertical membrane in this group include ABL, MLS, and ChAc.
protein interactions, which results in loss of membrane, decrease PNH is due to an acquired stem cell defect that results in the lack
in surface areatovolume ratio, and formation of spherocytes of GPI-anchored proteins on blood cell surfaces. CD55 and CD59,
that are destroyed in the spleen. Patients with HS have anemia, complement-regulating proteins, are partially or completely defi-
splenomegaly, jaundice, an increased MCHC, a negative DAT cient on RBCs, which makes the RBCs susceptible to spontaneous
result, biochemical evidence of hemolysis, and spherocytes and complement lysis. Flow cytometry is a sensitive method to detect
polychromasia on the peripheral blood film. Results on the osmotic the absence of GPI-anchored proteins on cell surfaces. The rate
fragility test are usually positive. of hemolysis in classic PNH improves after treatment with eculi-
HE and HPP are caused by mutations that disrupt the horizontal zumab, a complement C5 inhibitor.
interactions in the protein skeleton, which results in loss of G6PD deficiency is the most common RBC enzymopathy, but the
mechanical stability of the membrane. Elliptocytes are present on vast majority of patients are asymptomatic. Patients with class II
the peripheral blood film. Only 10% of HE patients have moderate and III G6PD variants may develop acute hemolytic anemia after
or severe anemia. HPP is a severe form of HE in which extreme infections or after ingestion of certain drugs or fava beans. A small
poikilocytosis as well as schistocytes, microspherocytes, and percentage of patients have class I G6PD variant and CNSHA.
elliptocytes are seen on the peripheral blood film. Most patients with PK deficiency have symptoms of hemolysis.
Hereditary ovalocytosis, also called SAO, is caused by a mutation Burr cells are commonly observed on the peripheral blood film. PK
in band 3 that increases membrane rigidity. The prevalence is high deficiency is the most common cause of CNSHA.
in Southeast Asia, and hemolysis is mild or absent; typical cells
are oval with one to two transverse bars or ridges. Now that you have completed this chapter, go back and
Hereditary stomatocytosis is a group of disorders characterized by read again the case study at the beginning and respond
an RBC membrane that leaks cations. In OHSt, the RBCs are to the questions presented.

R E V I E W Q UESTIONS
1. In HS a characteristic abnormality in the CBC results is: 2. The altered shape of the spherocyte in HS is due to:
a. Increased MCV a. An abnormal RBC membrane protein affecting
b. Increased MCHC vertical protein interactions
c. Decreased MCH b. Defective RNA synthesis
d. Decreased platelet and WBC count c. An extrinsic factor in the plasma
d. Abnormality in the globin composition of the
hemoglobin molecule
334 PART IV Hematopathology: Erythrocyte Disorders

3. Which of the following results are consistent with HS? 8. The most common manifestation of G6PD deficiency is:
a. Increased osmotic fragility, negative DAT result a. Chronic hemolytic anemia caused by cell shape
b. Decreased osmotic fragility, positive DAT result change
c. Increased osmotic fragility, positive DAT result b. Acute hemolytic anemia caused by drug exposure or
d. Decreased osmotic fragility, negative DAT result infections
c. Mild compensated hemolysis caused by ATP
4. The RBCs in HE are abnormally shaped and have unstable deficiency
cell membranes as a result of: d. Chronic hemolytic anemia caused by intravascular
a. Abnormal shear stresses in the circulation RBC lysis
b. Defects in horizontal membrane protein interactions
c. Mutations in ankyrin 9. A patient experiences an episode of acute intravascular
d. Lack of all Rh antigens in the RBC membrane hemolysis after taking an antibiotic for the first time. The
physician suspects that the patient may have G6PD defi-
5. The peripheral blood film for patients with mild HE is ciency and orders an RBC G6PD assay 2 days after the
characterized by: hemolytic episode begins. How will this affect the test
a. Elliptical RBCs result?
b. Oval RBCs with one or two transverse ridges a. No effect
c. Overhydrated RBCs with oval central pallor b. False increase due to reticulocytosis
d. Densely stained RBCs with a few irregular c. False decrease due to hemoglobinemia
projections d. Absence of enzyme activity

6. Laboratory test results for patients with HPP include all of 10. The most common defect or deficiency in the anaerobic
the following except: glycolytic pathway that causes CNSHA is:
a. RBCs that show marked thermal sensitivity at 41 C a. PK deficiency
to 45 C b. Lactate dehydrogenase deficiency
b. Marked poikilocytosis with elliptocytes, RBC c. G6PD deficiency
fragments, and microspherocytes d. Methemoglobin reductase deficiency
c. Low fluorescence when incubated with
eosin-5-maleimide 11. Which of the following laboratory tests would be best to
d. Increased MCV and normal RDW confirm PNH?
a. Acidified serum test (Ham test)
7. Acanthocytes are found in association with: b. Osmotic fragility test
a. Abetalipoproteinemia c. Flow cytometry for CD55 and CD59 on granulocytes
b. G6PD deficiency d. Prussian blue staining for iron in urine sediment
c. Rh deficiency syndrome
d. Vitamin B12 deficiency

REFERENCES
1. Mohandas N, Evans E: Mechanical properties of the red cell 6. Bennett V: Proteins involved in membrane-cytoskeleton asso-
membrane in relation to molecular structure and genetic ciation in human erythrocytes: spectrin, ankyrin, and band
defects. Annu Rev Biophys Biomol Struct 23:787-818, 1994. 3. Methods Enzymol 96:313-324, 1983.
2. Mohandas N, Chasis JA: Red blood cell deformability, mem- 7. Nicolas V, Le Van Kim C, Gane P, et al: Rh-RhAG/ankyrin-R,
brane material properties and shape: regulation by trans- a new interaction site between the membrane bilayer and the
membrane, skeletal and cytosolic proteins and lipids. Semin red cell skeleton, is impaired by Rhnull-associated mutation.
Hematol 30:171-192, 1993. J Biol Chem 278:25526-25533, 2003.
3. Mohandas N, Gallagher PG: Red cell membrane: past, 8. Salomao M, Zhang X, Yang Y, et al: Protein 4.1R-dependent
present, future. Blood 112:3939-3948, 2008. multiprotein complex: new insights into the structural orga-
4. Gallagher PG: The red blood cell membrane and its disorders: nization of the red blood cell membrane. Proc Natl Acad Sci
hereditary spherocytosis, elliptocytosis, and related diseases. U S A 105:8026-8031, 2008.
In Kaushansky K, Lichtman MA, Beutler T, et al, editors: 9. An X, Debnath G, Guo X, et al: Identification and functional
Williams hematology, ed 8, New York, 2010, McGraw-Hill characterization of protein 4.1R and actin-binding sites
Medical, chap 45. Available at: http://www.accessmedicine. in erythrocyte beta-spectrin: regulation of the interactions
com/content.aspx?aID=6244171. Accessed August 18, 2010. by phosphatidylinositol-4,5-biphosphate. Biochemistry 44:
5. Palek J, Jarolim P: Clinical expression and laboratory detec- 10681-10688, 2005.
tion of red blood cell membrane protein mutations. Semin 10. An X, Mohandas N: Disorders of red cell membrane. Br J
Hematol 30:249-283, 1993. Haematol 141:367-375, 2008.
CHAPTER 23 Intrinsic Defects Leading to Increased Erythrocyte Destruction 335
11. An X, Guo X, Zhang X, et al: Conformational stabilities of the 32. Bruce LJ: Hereditary stomatocytosis and cation leaky red
structural repeats of erythroid spectrin and their functional cellsrecent developments. Blood Cells Mol Dis 42:216-222,
implications. J Biol Chem 281:10527-10532, 2006. 2009.
12. Delaunay J: The molecular basis of hereditary red cell mem- 33. Bruce LJ, Guizouam H, Burton NM, et al: The monovalent
brane disorders. Blood Rev 21:1-20, 2007. cation leak in overhydrated stomatocytic red blood cells
13. Gallagher PG, Jarolim P: Red blood cell membrane disorders. results from amino acid substitutions in the Rh-associated
In Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology: glycoprotein. Blood 113:1350-1357, 2009.
basic principles and practice, ed 5, Philadelphia, 2009, Churchill 34. Carella M, Stewart G, Ajetunmobi JF, et al: Genomewide
Livingstone. search for dehydrated hereditary stomatocytosis (hereditary
14. Perrotta S, Gallagher PG, Mohandas N: Hereditary spherocy- xerocytosis): mapping of locus to chromosome 16 (16q23-
tosis. Lancet 372:1411-1426, 2008. qter). Am J Hum Genet 63:810-816, 1998.
15. Bolton-Maggs PHB, Stevens RF, Dodd NJ, et al: Guidelines for 35. Rampoldi L, Danek A, Monaco AP: Clinical features and
the diagnosis and management of hereditary spherocytosis. molecular bases of neuroacanthocytosis. J Mol Med 80:475-
Br J Haematol 126:455-474, 2004. 491, 2002.
16. De Franceschi L, Olivieri O, Miraglia del Giudice E, et al: 36. Walker RH, Jung HH, Dobson-Stone C, et al: Neurologic
Membrane cation and anion transport activities in erythro- phenotypes associated with acanthocytosis. Neurology 68:92-
cytes of hereditary spherocytosis: effects of different mem- 98, 2007.
brane protein defects. Am J Hematol 55:121-128, 1997. 37. Parker CJ: Paroxysmal Nocturnal Hemoglobinuria. In
17. Hassoun H, Vassiliadis JN, Murray J, et al: Characterization Kaushansky K, Lichtman MA, Beutler TJ, et al, editors:
of the underlying molecular defect in hereditary spherocyto- Williams hematology, ed 8, New York, 2010, McGraw-Hill
sis associated with spectrin deficiency. Blood 90:398-406, Medical, chap 40. Available at: http://www.accessmedicine.
1997. com/content.aspx?aID=6109660. Accessed August 18, 2010.
18. Dhermy D, Galand C, Bournier O, et al: Heterogenous band 38. Besslar M, Hiken J: The pathophysiology of disease in patients
3 deficiency in hereditary spherocytosis related to different with paroxysmal nocturnal hemoglobinuria. Hematology Am
band 3 gene defects. Br J Haematol 98:32-40, 1997. Soc Hematol Educ Program, pp 104-110, 2008.
19. Bouhassira EE, Schwartz RS, Yawata Y, et al: An alanine-to- 39. Brodsky RA: Paroxysmal nocturnal hemoglobinuria. In
threonine substitution in protein 4.2 cDNA is associated with Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology:
a Japanese form of hereditary hemolytic anemia (protein basic principles and practice, ed 5, Philadelphia, 2009, Churchill
4.2NIPPON). Blood 79:1846-1854, 1992. Livingstone.
20. Ballas SK, Clark MR, Mohandas N, et al: Red cell membrane 40. Takeda J, Miyata T, Kawagoe K, et al: Deficiency of the GPI
and cation deficiency in Rh null syndrome. Blood 63:1046- anchor caused by a somatic mutation of the PIG-A gene
1055, 1984. in paroxysmal nocturnal hemoglobinuria. Cell 73:703-711,
21. Clark MR, Mohandas N, Shohet SB: Osmotic gradient ekta- 1993.
cytometry: comprehensive characterization of red cell volume 41. Parker C, Omine M, Richards S, et al: Diagnosis and manage-
and surface maintenance. Blood 61:899-910, 1983. ment of paroxysmal nocturnal hemoglobinuria. Blood 106:
22. King M-J, Smythe J, Mushens R: Eosin-5-maleimide binding 3699-3709, 2005.
to band 3 and Rh-related proteins forms the basis of a screen- 42. Madkaikar M, Gupta M, Jijina F, et al: Paroxysmal nocturnal
ing test for hereditary spherocytosis. Br J Haematol 124:106- haemoglobinuria: diagnostic tests, advantages, and limita-
113, 2004. tions. Eur J Haematol 83:503-511, 2009.
23. Streichman S, Gescheidt Y: Cryohemolysis for the detection 43. Sutherland DR, Kuek N, Azcona-Olivera J, et al: Use of
of hereditary spherocytosis: correlation studies with osmotic FLAER-based WBC assay in the primary screening of PNH
fragility and autohemolysis. Am J Hematol 58:206-212, 1998. clones. Am J Clin Pathol 132:564-572, 2009.
24. Abdullah F, Zhang Y, Camp M, et al: Splenectomy in heredi- 44. Dmytrijuk A, Robie-Suh K, Cohen MH, et al: FDA report:
tary spherocytosis: review of 1,657 patients and application eculizumab (Soliris) for the treatment of patients with par-
of the pediatric quality indicators. Pediatr Blood Cancer oxysmal nocturnal hemoglobinuria. Oncologist 13:993-1000,
52:834-837, 2009. 2008.
25. Gallagher PG: Hereditary elliptocytosis: spectrin and protein 45. Gregg XT, Prchal JT: Red blood cell enzymopathies. In
4.1R. Semin Hematol 41:142-164, 2004. Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology:
26. Dhermy D, Schrevel J, Lecomte M-C: Spectrin-based skeleton basic principles and practice, ed 5, Philadelphia, 2009, Churchill
in red blood cells and malaria. Curr Opin Hematol 14:198- Livingstone.
202, 2007. 46. Cappellini MD, Fiorelli G: Glucose-6-phosphate dehydroge-
27. Winardi R, Reid M, Conboy J, et al: Molecular analysis nase deficiency. Lancet 371:64-74, 2008.
of glycophorin C deficiency in human erythrocytes. Blood 47. van Solinge Wouter W, van Wijk R: Disorders of Red Cells
81:2799-2803, 1993. Resulting from Enzyme Abnormalities. In Kaushansky K,
28. King M-J, Telfer P, MacKinnon H, et al: Using the eosin-5- Lichtman MA, Beutler TJ, et al, editors: Williams hematology,
maleimide binding test in the differential diagnosis of ed 8, New York, 2010, McGraw-Hill Medical, chap 46. Avail-
hereditary spherocytosis and hereditary pyropoikilocytosis. able at: http://www.accessmedicine.com/content.aspx?aID=
Cytometry B Clin Cytom 74B:244-250, 2008. 6120127. Accessed August 19, 2010.
29. Mohandas N, Winardi R, Knowles D, et al: Molecular basis 48. Nkhoma ET, Poole C, Vannappagari V, et al: The global preva-
for membrane rigidity of hereditary ovalocytosis. A novel lence of glucose-6-phosphate dehydrogenase deficiency: a
mechanism involving the cytoplasmic domain of band 3. systematic review and meta-analysis. Blood Cells Mol Dis
J Clin Invest 89:686-692, 1992. 42:267-278, 2009.
30. Liu S-C, Zhai S, Palek J, et al: Molecular defect of the band 3 49. Luzzatto L: Glucose 6-phosphate dehydrogenase deficiency:
protein in Southeast Asian ovalocytosis. N Engl J Med from genotype to phenotype. Hematologica 91:1303-1306,
323:1530-1538, 1990. 2006.
31. Schofield AE, Tanner MJA, Pinder JC, et al: Basis of unique 50. Guindo A, Fairhurst RM, Doumbo OK, et al: X-linked G6PD
red cell membrane properties in hereditary ovalocytosis. deficiency protects hemizygous males but not heterozygous
J Mol Biol 223:949-958, 1992. females against severe malaria. PLoS 4(3):e66, 2007.
336 PART IV Hematopathology: Erythrocyte Disorders

51. Leslie T, Briceno M, Mayan I, et al: The impact of phenotypic 56. Kanno H, Ballas SK, Miwa S, et al: Molecular abnormality of
and genotypic G6PD deficiency on risk of Plasmodium vivax erythrocyte pyruvate kinase deficiency in the Amish. Blood
infection: a case-control study amongst Afghan refugees in 83:2311-2316, 1994.
Pakistan. PLoS 7(5):e1000283, 2010. 57. Zanella A, Fermo E, Bianchi P, et al: Red cell pyruvate kinase
52. Glucose-6-phosphate dehydrogenase deficiency. WHO deficiency: molecular and clinical aspects. Br J Haematol
Working Group. Bull World Health Organ 67:601-611, 130:11-25, 2005.
1989. 58. Van Wijk R, van Solinge WW: The energy-less red blood cell
53. Youngster I, Arcavi L, Schechmaster R, et al: Medications and is lost: erythrocyte enzyme abnormalities of glycolysis. Blood
glucose-6-phosphate dehydrogenase deficiency: an evidence- 106:4034-4042, 2005.
based review. Drug Saf 33:713-726, 2010. 59. Andersen FD, dAmore F, Nielsen FC, et al: Unexpectedly
54. Prchal JT, Gregg XT: Red cell enzymes. Hematol Am Soc high but still asymptomatic iron overload in a patient with
Hematol Educ Program, pp 19-23, 2005. pyruvate kinase deficiency. Hematol J 5:543-545, 2004.
55. Beutler E, Gelbart T: Estimating the prevalence of pyruvate 60. Tsang SS, Feng CS: A modified screening procedure to detect
kinase deficiency from the gene frequency in the general pyruvate kinase deficiency. Am J Clin Pathol 99:128-131,
white population. Blood 95:3585-3588, 2000. 1993.
Extrinsic Defects Leading to
Increased Erythrocyte Destruction
24
Nonimmune Causes
Elaine M. Keohane

OUTLINE OBJECTIVES
Microangiopathic After completion of this chapter, the reader will be able to:
Hemolytic Anemia
Thrombotic 1. Describe the general pathophysiology and clinical characteristic morphology on a peripheral blood film,
Thrombocytopenic laboratory findings in microangiopathic hemolytic the extent of parasitemia, and the length of the
Purpura anemia, including the characteristic red blood cell erythrocytic cycle.
Hemolytic Uremic morphology. 7. Describe the proper specimen collection and proce-
Syndrome 2. Compare and contrast the pathophysiology, clinical dure for performing a thin and thick blood film
HELLP Syndrome symptoms, and typical laboratory findings in throm- examination.
Disseminated botic thrombocytopenic purpura, hemolytic uremic 8. Compare and contrast Babesia species and Plasmo-
Intravascular syndrome, HELLP (hemolysis, elevated liver enzymes, dium species in terms of geographic distribution,
Coagulation and low platelet count) syndrome, and disseminated clinical symptoms of infection, and morphology.
Hypertensive Crisis
intravascular coagulation. 9. Describe the pathophysiology, laboratory findings,
Macroangiopathic
3. Explain the pathophysiology and typical laboratory fea- and peripheral blood morphology in hemolytic
Hemolytic Anemia
Traumatic Cardiac tures of hypertensive crises, traumatic cardiac hemo- anemia due to clostridial sepsis, bartonellosis, drugs,
Hemolytic Anemia lytic anemia, and exercise-induced hemoglobinuria. chemicals, venoms, and extensive burns.
Exercise-Induced 4. Describe the life cycle of Plasmodium, including the 10. Given the history, symptoms, laboratory findings,
Hemoglobinuria hepatic and erythrocytic cycles, and the insect vector. and a representative microscopic field from a periph-
Hemolytic Anemia Caused 5. Explain the pathophysiologic mechanisms in P. fal- eral blood film of a patient with suspected extrinsic,
by Infectious Agents ciparum infection that lead to anemia and neurologic nonimmune hemolytic anemia, discuss possible
Malaria manifestations. causes of the anemia and indicate the data that
Babesiosis 6. Differentiate the five Plasmodium species affecting support the conclusions.
Clostridial Sepsis humans based on the geographic distribution,
Bartonellosis
Hemolytic Anemia Caused
by Other Red Blood Cell CASE STUDY
Injury After studying the material in this chapter, the reader should be able to respond to the following
Drugs and Chemicals case study:
Venoms
Extensive Burns (Thermal A 24-year-old woman was brought to the emergency department with a 2-day history of fever,
Injury) chills, excessive sweating, nausea, and general malaise. Because she had recently returned from a
3-week family trip to Ghana in Western Africa, the treating physician ordered a CBC and examina-
tion of thin and thick peripheral blood films. The following are the patients laboratory results:
Patient Results Reference Range
9
WBCs (10 /L) 11.0 4.5-11.0
Hb (g/dL) 8.7 12.0-15.0
Hct (%) 25 35-49
MCV (fL) 92 80-100
Platelets (109/L) 176 150-450
Continued

337
338 PART IV Hematopathology: Erythrocyte Disorders

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

Figure 24-2 Thick peripheral blood film for the patient in the case study
Figure 24-1 Thin peripheral blood film for the patient in the case study (1000). (From Marler LM, etal: Indiana Pathology Images, Indianapolis,
(1000). Ind.)

Inclusions were noted on the thin and thick peripheral 2. What is the likely diagnosis for this patient?
blood films (Figures 24-1 and 24-2). 3. What clues in the history support this diagnosis?
Based on the results of the CBC and peripheral blood 4. What other forms might be found on the peripheral blood
films, the patient was treated with oral quinine sulfate and films in this disease?
doxycycline. 5. What are the pathophysiologic mechanisms for the anemia
1. Identify the inclusions present on the thin and thick in this disease?
peripheral blood films.

E
xtrinsic hemolytic anemias comprise a diverse group of
disorders in which red blood cells (RBCs) are structur- BOX 24-1 Extrinsic Conditions Causing Nonimmune
ally and functionally normal, but a condition outside of Red Blood Cell (RBC) Injury and Hemolytic Anemia
the RBCs causes premature hemolysis. The extrinsic hemolytic Microangiopathic hemolytic anemia
anemias can be divided into conditions with nonimmune and Thrombotic thrombocytopenic purpura
immune causes. A common feature in the nonimmune extrinsic Hemolytic uremic syndrome
hemolytic anemias is the presence of a condition that causes HELLP syndrome
physical or mechanical injury to the RBCs. This injury can be Disseminated intravascular coagulation
caused by abnormalities in the microvasculature (microangio- Hypertensive crisis
pathic) or the heart and large blood vessels (macroangiopathic), Macroangiopathic hemolytic anemia
infectious agents, chemicals, drugs, venoms, or extensive burns. Traumatic cardiac hemolytic anemia
The nonimmune disorders causing hemolytic anemia are dis- Exercise-induced hemoglobinuria
cussed in this chapter and are summarized in Box 24-1. In Infection
disorders in the immune category, the hemolytic anemia is Malaria
caused by antibodies, complement, or both, and these condi- Babesiosis
tions are covered in Chapter 25. Examination of a peripheral Clostridial sepsis
blood film is important in suspected extrinsic hemolytic Bartonellosis
anemias, because observation of abnormal RBC morphology, RBC injury due to other causes
such as schistocytes, spherocytes, or the presence of intracel- Chemicals
lular organisms, provides an important clue to the diagnosis. Drugs
Venoms
Extensive burns
MICROANGIOPATHIC HEMOLYTIC ANEMIA
HELLP, Hemolysis, elevated l iver enzymes, and low platelet count.
Microangiopathic hemolytic anemias (MAHAs) are a group of
disorders characterized by RBC fragmentation, hemolysis, and
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 339

anemia. The fragmentation occurs intravascularly by the different etiologies and require different treatments.5 This
mechanical shearing of RBC membranes as the cells rapidly chapter provides a summary of these conditions. TTP, HUS,
pass through turbulent areas of small vessels that are partially and HELLP syndrome are discussed in more detail in Chapter
blocked by microthrombi or damaged endothelium.1,2 Upon 43. DIC is discussed in more detail in Chapter 42.
shearing, RBC membranes quickly reseal with minimal escape
of hemoglobin (Hb), but the resulting fragments (called schisto- Thrombotic Thrombocytopenic Purpura
cytes) are distorted and become rigid.1 The spleen clears the rigid TTP is a rare disorder characterized by the abrupt appearance
RBC fragments from the circulation through the extravascular of microangiopathic hemolytic anemia, severe thrombocytope-
hemolytic process (see Chapter 22).1 Laboratory evidence of the nia, and a markedly elevated level of serum lactate dehydroge-
hemolytic anemia includes a decreased hemoglobin and hema- nase.2,6 Neurologic dysfunction, fever, and renal failure may
tocrit (Hct), reticulocytosis, increased serum unconjugated also occur, but they are not consistently present.2 TTP is more
bilirubin, decreased serum haptoglobin, and increased urine commonly found in adults and is rare in children, and there
urobilinogen. In some cases, the fragmentation is so severe that is a higher incidence in females than in males.6,7 Typical labora-
intravascular hemolysis occurs with varying amounts of hemo- tory findings at presentation include a hemoglobin concentra-
globinemia, hemoglobinuria, and markedly decreased levels tion of less than 10g/dL, a platelet count of less than 20
of serum haptoglobin.1 The presence of schistocytes on the 109/L, and schistocytes observed on the peripheral blood film
peripheral blood film is a characteristic feature of microangio- (Figure 24-3). After the bone marrow begins to respond to the
pathic hemolytic anemia. The RBC shearing may also produce anemia, polychromasia and nucleated RBCs may also be
helmet cells and, occasionally, microspherocytes. Polychroma- observed. The white blood cell (WBC) count is often increased,
sia and nucleated RBCs are found when the anemia is severe. and immature granulocytes (left shift) may appear. The bone
Thrombotic microangiopathy is a more specific term used to marrow shows erythroid hyperplasia and a normal number of
describe disorders in which the microangiopathic hemolytic megakaryocytes. Various amounts of protein, RBCs, and
anemia is due to the formation of thrombi in the microvascu- urinary casts may be present in the urine depending on the
lature. In addition to the fragmentation hemolysis, thrombo- extent of the renal damage. Results of coagulation tests are
cytopenia and ischemic organ injury are characteristic findings. usually within the reference range, which differentiates TTP
The thrombotic microangiopathies include thrombotic throm- from DIC.
bocytopenic purpura (TTP), hemolytic uremic syndrome The pathophysiology of TTP involves the release of long
(HUS), and HELLP (hemolysis, elevated liver enzymes, low von Willebrand factor (VWF) multimers from vascular endo-
platelet count) syndrome.3 Disseminated intravascular coagula- thelial cells.8 The long VWF multimers adhere to endothelial
tion (DIC) can also be classified as a thrombotic micro cells and rapidly bind platelets through specific receptor-ligand
angiopathy; however, schistocytes are found in the peripheral interaction and subsequently trigger platelet aggregation. In the
blood film in only about half of the cases.4 TTP and HUS may process platelets are removed from circulation, which causes
be difficult to differentiate, because they can have overlapping severe thrombocytopenia, and the platelet-VWF (hyaline)
clinical and laboratory findings (Box 24-2); however, they have microthrombi block small blood vessels, which leads to isch-
emia in the brain, kidney, and other organs.2,8 In addition,
RBCs are sheared as they pass through small vessels partially
blocked by microthrombi, which results in RBC fragmenta-
BOX 24-2 Typical Laboratory Findings in TTP tion and hemolytic anemia. The marked increase in lactate
and HUS
Hematologic
Decreased hemoglobin (<10g/dL)
Decreased platelets
Increased reticulocyte count
Peripheral blood film
Schistocytes
Polychromasia
Nucleated red blood cells (severe cases)
Biochemical
Markedly increased lactate dehydrogenase*
Increased serum total and unconjugated bilirubin
Decreased haptoglobin
Hemoglobinemia
Hemoglobinuria
Proteinuria, hematuria, casts

HUS, Hemolytic uremic syndrome; TTP, thrombotic thrombocytopenic purpura. Figure 24-3 Peripheral blood film from a patient with thrombotic throm-
*From systemic ischemia and hemolysis; more commonly found in TTP. bocytopenic purpura. Note the schistocytes and a nucleated red blood cell

From acute renal failure; more commonly found in HUS. (1000).


340 PART IV Hematopathology: Erythrocyte Disorders

dehydrogenase is attributed more to the massive tissue isch- of serum creatinine, proteinuria, hematuria, and the presence
emia than to the RBC lysis. of hyaline, granular, and RBC casts in the urine (see Box 24-2).
TTP may be acquired or familial. The acquired type is very The D+ HUS type comprises 90% of cases of HUS.13 The
heterogeneous, and the mechanisms that trigger the TTP patho- most common cause is infection with enterohemorrhagic sero-
physiology are not completely clear. Approximately half of the types of Escherichia coli (such as O157:H7).3 D+ HUS occurs
patients present with secondary TTP associated with stem cell most often in young children, but can be found in patients of
transplantation, pregnancy, disseminated cancer, autoimmune all ages. Patients initially have acute gastroenteritis, often with
disorders, systemic infection, and use of certain drugs, includ- bloody diarrhea, and after 1 to 3 days develop oliguria and
ing antiplatelet and chemotherapeutic agents.3,6,7 Of the other symptoms of renal impairment. About one fourth of
remaining nonsecondary or idiopathic TTP cases, about 50% patients develop neurologic manifestations.13
to 80% show a severe deficiency (less than 10% activity) of the E. coli serotypes implicated in HUS release Shiga toxins
VWF-cleaving protease known as a disintegrin-like and metallo- (Stx-1 and Stx-2) that are absorbed from the intestines into the
protease domain with thrombospondin type 1 motifs 13 (ADAMTS plasma, and have an affinity for the Gb3 glycolipid receptors
13).6,7,9 Under normal conditions, ADAMTS 13 cleaves the long on endothelial cells, particularly those in the kidney, brain,
multimers of VWF into shorter segments as they are released and intestines.3,14 The toxin is transported into the endothelial
from the endothelial cells, thus preventing excessive platelet cells, where it inhibits protein synthesis and causes major
adhesion, aggregation, and microthrombi formation.2,5 The endothelial cell injury. Although the mechanisms are not
ADAMTS 13 deficiency in acquired TTP has been attributed to known, the Shiga toxin, together with many cytokines secreted
inhibition by an immunoglobulin G (IgG) autoantibody, but as a result of the infection, may also induce changes in endo-
inhibitory autoantibodies are not consistently found in all thelial cells that are prothrombotic, including expression of
patients with the deficiency.6,10,11 For the remaining cases of adhesion molecules and secretion of increased amounts of
idiopathic TTP, the underlying cause is not known.9 long VWF multimers.3,14 The end result is endothelial cell dis-
Approximately 80% of patients with idiopathic TTP respond ruption, formation of platelet-fibrin thrombi that block the
favorably to plasma exchange therapy, likely due to the removal microvasculature in the glomeruli, and acute renal failure.3,14
of the offending ADAMTS 13 autoantibody and infusion of Microthrombi can also form in other organs in addition to the
replacement ADAMTS 13 enzyme from donor plasma.5,7 There- kidney. There is no specific treatment for D+ HUS, but patients
fore it is important that this type of TTP be recognized and are provided supportive care as needed, including hydration,
quickly treated with plasma exchange therapy to avoid a fatal dialysis, and transfusions.13,14 The symptoms usually resolve
outcome.5 Approximately one third of patients who respond spontaneously in 1 to 3 weeks, and the prognosis is favorable
to plasma exchange experience recurrent episodes of TTP.3 for most patients.13
Except when the TTP is related to autoimmune disease, preg- Atypical HUS comprises about 10% of cases of HUS, is
nancy, or ticlopidine use, patients with secondary TTP gener- more commonly found in adults, and has a poor prognosis.12,15
ally do not respond well to plasma exchange, and the prognosis Approximately half of the cases are familial and the other half
in these cases is poor.5,6,7,12 are sporadic. The familial type is caused by mutations in genes
Familial TTPs are rare disorders due to mutations in the that code for proteins involved in the alternative complement
ADAMTS13 gene. Over 90 different mutations have been iden- pathway or its regulation, such as the genes for factor H, factor
tified, and symptomatic individuals are either homozygous for I, factor B, membrane cofactor protein, and thrombomodu-
one of the mutations or compound heterozygous for two dif- lin.12,15 The mutations result in uncontrolled activation of the
ferent mutations.12 The mutations result in reduced ADAMTS complement pathway, which causes endothelial injury, activa-
13 activity, usually 5% to 10% of normal.3 Familial TTP usually tion of platelets and coagulation factors, and formation of
presents in infancy with recurrent episodes; however, some platelet-fibrin thrombi that obstruct the glomerular microvas-
patients may not be symptomatic until adulthood after their culature.3,16 The sporadic form occurs secondary to some drugs,
system is stressed by pregnancy or a severe infection. Infusion cancer, organ transplantation, pregnancy, and human immu-
of fresh frozen plasma supplies the deficient ADAMTS 13 nodeficiency virus (HIV) infection.16
enzyme.3
HELLP Syndrome
Hemolytic Uremic Syndrome HELLP syndrome is a serious complication in pregnancy and
HUS is characterized by microangiopathic hemolytic anemia, is named for its characteristic presentation of hemolysis, ele-
thrombocytopenia, and acute renal failure from thrombi vated liver enzymes, and low platelet count. It occurs in fewer
formed mainly in the glomerular microvasculature.5 There are than 1% of all pregnancies, but develops in approximately
two general types of HUS: D+ HUS, which is preceded by an 10% to 20% of patients with preeclampsia, most often in the
episode of acute gastroenteritis, often with bloody diarrhea; third trimester.17 The exact pathogenesis is not known. In
and atypical HUS, which does not have antecedent diarrhea.5 preeclampsia, abnormalities in the development of placental
Patients with HUS have the typical laboratory findings of vasculature results in poor perfusion and hypoxia. As a
microangiopathic hemolytic anemia discussed previously. In result, antiangiogenic proteins are released from the placenta
addition, there is usually a mild to moderate decrease in plate- that block the action of placental growth factors, including
lets and evidence of renal failure, including an elevated level vascular endothelial growth factor.3,18 The continued vascular
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 341

insufficiency of the placenta results in maternal endothelial cell brought under control, the RBC fragmentation disappears and
dysfunction, which leads to platelet activation and fibrin depo- the platelet count returns to normal within a few days.22,23
sition in the microvasculature, particularly in the liver.18
Anemia, biochemical evidence of hemolysis, and schisto-
MACROANGIOPATHIC HEMOLYTIC ANEMIA
cytes on the peripheral blood film are found as in the other
thrombotic microangiopathies. The platelet count is less than Traumatic Cardiac Hemolytic Anemia
100 109/L; counts falling below 50 109/L indicate a worse Mechanical hemolysis can occur in patients with prosthetic
prognosis.18,19 The level of lactate dehydrogenase is elevated, cardiac valves due to the turbulent blood flow through and
which reflects the hepatic necrosis as well as the hemolysis. The around the implanted device.18,24 The hemolysis is usually
level of aspartate aminotransferase can be markedly elevated mild, and anemia does not generally develop due to compen-
due to the severe hepatocyte injury. The low platelet count and sation by the bone marrow.24 Severe hemolysis is rare and is
increased levels of lactate dehydrogenase and aspartate amino- usually due to paravalvular leaks in prosthetic cardiac valves.24
transferase are major diagnostic criteria for the HELLP syn- Hemolysis can also occur in patients with cardiac valve disease
drome and are used to assess the severity of the disease.17,18 The prior to corrective surgery.18 The anemia that occurs in severe
prothrombin time and the partial thromboplastin time are cases is usually normocytic, but can be microcytic if iron defi-
within the reference range, which distinguishes the HELLP syn- ciency develops due to chronic urinary hemoglobin loss.
drome from DIC. Therapy includes delivery of the fetus and Depending on the severity of the hemolysis and the ability
placenta as soon as possible along with supportive care to of the bone marrow to compensate for the reduced RBC life
control seizures, hypertension, and fluid balance. The mortality span, patients can be asymptomatic or present with pallor,
rate is 3% to 5% for the mother and 9% to 24% for the fetus.18 fatigue, and even heart failure.24 On the peripheral blood film,
schistocytes are a characteristic feature due to the mechanical
Disseminated Intravascular Coagulation fragmentation of the RBCs (Figure 24-4). The reticulocyte
DIC is characterized by the widespread activation of the hemo- count is increased, but the platelet count is within the reference
static system, resulting in fibrin thrombi formation throughout range. Lactate dehydrogenase activity and levels of serum indi-
the microvasculature. The major clinical manifestations are rect bilirubin and plasma hemoglobin are elevated, whereas
organ damage due to obstruction of the microvasculature and the serum haptoglobin level is decreased. Hemoglobinuria
bleeding due to the consumption of platelets and coagulation may be observed in severe hemolysis. Hemosiderinuria and a
factors and secondary activation of fibrinolysis.20 DIC is a decreased level of serum ferritin occur with chronic hemoglo-
complication of many disorders, such as disseminated cancers, binuria due to the urinary loss of iron.
acute leukemias, infections, obstetric complications, crush or Surgical repair or replacement of the prothesis may be
brain injuries, acute hemolytic transfusion reactions, extensive required if the anemia is severe enough to require transfusions.
burns, snake or spider envenomation, and chronic inflamma- For patients with hemoglobinuria, iron supplementation is
tion (see Table 42-14). provided to replace urinary iron loss. Folic acid may also be
Thrombocytopenia of varying degree is a consistent finding. required, because deficiencies can occur due to the increased
Only about half the patients have schistocytes on the periph- erythropoietic activity in the bone marrow.24
eral blood film.4 The prothrombin time and partial thrombo-
plastin time are prolonged, the fibrinogen level is decreased, Exercise-Induced Hemoglobinuria
and the level of D-dimer is increased in DIC, which distinguish RBC hemolysis, with an increase in free plasma hemoglobin
it from the other thrombotic microangiopathies. and a decrease in serum haptoglobin level, has been

Hypertensive Crisis
Hypertensive crisis or hypertensive emergency (previously
called malignant hypertension) is a severe and abrupt increase in
blood pressure with acute organ damage, particularly in the
heart, brain, and kidney.21 The sudden mechanical stress
induced by the elevated blood pressure damages the endothe-
lial cells and results in increased vascular permeability, activa-
tion of coagulation factors and platelets, and fibrin deposition
in damaged vessel walls.21 The manifestations of hypertensive
crisis include encephalopathy, acute myocardial infarction,
aortic dissection, pulmonary edema, severe preeclampsia,
acute renal failure, and microangiopathic hemolytic anemia.21,22
In one study, 27% of patients with hypertensive crisis had
microangiopathic hemolytic anemia when the criteria were a
platelet count of less than 150 109/L and either an elevated
level of lactate dehydrogenase or the presence of schistocytes Figure 24-4 Peripheral blood film for a patient with traumatic cardiac
on the peripheral blood film.22 When the hypertension is hemolytic anemia. Note the presence of schistocytes (1000).
342 PART IV Hematopathology: Erythrocyte Disorders

demonstrated after long-distance running and to a lesser extent disease.36 Greater than a 50% reduction in deaths due to
after intensive cycling and swimming,25-27 but frank hemoglo- malaria has been realized in some countries where malaria is
binuria after exercise is a rare occurrence.28 Exercise-induced endemic, and the goal is to achieve near zero worldwide deaths
hemoglobinuria has been reported mainly in endurance from malaria by 2015.36
runners, but has also been observed after strenuous hand From 1985 to 2008, the United States had an annual average
drumming.25,29 Various causes have been proposed, including of 1400 cases and five deaths from malaria, mostly in individu-
mechanical trauma from the forceful, repeated impact of the als returning from visits to areas in which it is endemic.38,39 P.
feet or hands on hard surfaces,25,26,29 increased RBC susceptibil- falciparum and P. vivax cause most of the infections seen in the
ity to oxidative stress,30,31 and exercise-induced alterations in United States.39
membrane cytoskeletal proteins.31,32
Exercise-induced hemoglobinuria does not usually cause Plasmodium Life Cycle
anemia unless the hemoglobinuria is particularly severe and Malaria is transmitted to humans by the bite of an infected
recurrent.28 Laboratory findings include a decreased level of female Anopheles mosquito. During a blood meal, sporozoites
serum haptoglobin, an elevated level of free plasma hemoglo- from the salivary gland of the mosquito are injected into the
bin, and hemoglobinuria observed after strenuous exercise. skin and migrate into the bloodstream of the human host. The
Patients also have a slight increase in mean cell volume (MCV) sporozoites rapidly leave the circulating blood and invade
and reticulocyte count, and with rare exceptions schistocytes hepatic parenchymal cells to begin exoerythrocytic schizogony,
are not observed on the peripheral blood film.28,29 Exercise- the parasites asexual cycle. After 5 to 16 days (depending on
induced hemoglobinuria is a diagnosis of exclusion, and other the species), hepatic cells rupture, with each cell releasing tens
possible causes of hemolysis and hemoglobinuria should be of thousands of merozoites into the bloodstream to invade
investigated and ruled out.28 There is no treatment for the circulating RBCs, thus beginning erythrocytic schizogony.40 Inside
disorder other than minimizing the physical impact on the feet the erythrocyte, the merozoite grows and metabolizes the
with padding in shoes, running on softer terrain, or, if hemo- hemoglobin. The merozoite becomes a ring form, which grows
lysis is severe, discontinuing the activity. into a mature trophozoite, then into an immature schizont
(chromatin dividing), and finally into a mature schizont that
contains merozoites. The merozoites are released from the
HEMOLYTIC ANEMIA CAUSED BY
erythrocytes into the bloodstream and invade other RBCs to
INFECTIOUS AGENTS
continue the asexual cycle. As the infection continues, the
Malaria cycles often recur at regular intervals as all the individual para-
Malaria is a potentially fatal condition caused by infection of sitic cycles become synchronous; this produces paroxysms of
RBCs with protozoan parasites of the genus Plasmodium. Most fever and chills at a frequency that varies according to malaria
human infections are caused by P. falciparum and P. vivax, but species. Resting stages of P. vivax and P. ovale, called hypnozoites,
P. ovale, P. malariae, and a fifth species, P. knowlesi, also infect can remain dormant in the liver and produce a relapse months
humans. P. knowlesi, a natural parasite of macaque monkeys, or years later.41,42
is easily misdiagnosed as P. malariae by microscopy, because Some merozoites enter RBCs and form male and female
the organisms are difficult to distinguish morphologically. gametocytes (sexual stages). Gametocytes are ingested by an
Since 2004, hundreds of microscopically identified cases of P. Anopheles mosquito when it takes a blood meal. The female
malariae infection in Malaysia were actually found to be caused gamete is fertilized by the male gamete in the mosquito gut to
by P. knowlesi when polymerase chain reaction (PCR) assays produce a zygote, which becomes an ookinete that migrates to
were used, including tests on archival blood films from the outer wall of the mosquito midgut and develops into an
1996.33-35 With the use of molecular techniques, P. knowlesi oocyst. The oocyst produces sporozoites that are released into
malaria is now known to be widespread in Malaysia, and cases the hemocele and migrate to the salivary glands of the mos-
have been reported in other areas of Southeast Asia.33-35 quito. When the mosquito takes a blood meal, the sporozoites
are inoculated into the human host.
Incidence Other modes of transmission include congenital infection
Half the worlds population (approximately 3.3 billion people) and transmission by blood transfusion, organ transplantation,
live in areas in which malaria is endemic and are at risk for the or sharing of syringes and needles, but malaria acquired
disease.36,37 Worldwide in 2008, there were an estimated 243 by these routes and local mosquito-transmitted malaria
million cases of malaria with 863,000 deaths, mostly in chil- contracted within this country occur infrequently in the
dren younger than 5 years of age.36 The majority of deaths United States.39,43
were in Africa (85%), followed by Southeast Asia (10%) and
the Eastern Mediterranean region (4%).36 The World Health Pathogenesis
Organization is coordinating a major global effort to control If an individual is bitten by infected mosquitos, the clinical
and eliminate malaria. These efforts include implementation outcome may be (1) no infection, (2) asymptomatic parasitemia
of indoor chemical spraying, distribution of millions of (the patient has no symptoms but parasites are present in the
insecticide-treated sleeping nets in high-risk areas, and promo- blood), (3) uncomplicated malaria (the patient has symptoms
tion of policies for appropriate treatment in regions of endemic and parasitemia but no organ dysfunction), or (4) severe
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 343
malaria (the patient has symptoms, parasitemia, and major delivery to organs and (2) protecting the parasite from clear-
organ dysfunction).36,41 The clinical outcome depends on para- ance by the spleen.40,41 In the placental microvasculature,
site factors (species, number of sporozoites injected, multi adherence of infected RBCs to endothelial cells results in local
plication rate, virulence, drug resistance), host factors (age, inflammation that can cause severe maternal anemia, decreased
pregnancy status, immune status, previous exposure, genetic fetal growth, premature delivery, and increased risk of fetal
polymorphisms, nutrition status, coinfection with other patho- loss.40,41 In the brain microvasculature, adherence of infected
gens, and duration of infection), geographic and social factors RBCs to endothelial cells can cause lethal cerebral malaria.
(endemicity, poverty, and availability of prompt and effective Infected RBC adherence is mediated by the binding of PfEMP1,
treatment), and other as yet unknown factors.40,41,44 In areas of a parasite protein expressed on the surface of infected RBCs, to
high Plasmodium transmission, most individuals develop cell receptors, particularly CD36 on platelets and some endo-
immunity, and the major risk groups for severe malaria are thelial cells.40,49 Adherence of infected RBCs in the brain has
children younger than 5 years of age and women in their first been attributed to a complex formed by VWF released by
pregnancy.45 Even in these risk groups, severe malaria is infre- cytokine-stimulated endothelial cells, platelets bound to the
quent.41 On the other hand, in residents of areas of low Plas- VWF, and infected RBCs bound to platelet CD36.50
modium transmission and in travelers to regions where malaria The ability of the parasite to invade RBCs affects the extent
is endemic, immunity is low or nonexistent, and individuals of the parasitemia and the severity of the disease. P. vivax and
of all ages are at risk for severe malaria.45 Most cases of severe P. ovale can only invade reticulocytes, and P. malariae can only
malaria are due to P. falciparum; however, P. vivax and P. invade older RBCs. On the other hand, P. falciparum and P.
knowlesi can also cause severe disease.34,40 Infection with P. knowlesi are able to invade RBCs of all ages and thus can lead
malariae or P. ovale, however, is usually uncomplicated and to very high levels of parasitemia.40,42 P. vivax requires Duffy
benign. Hyperparasitemia, defined as greater than 2% to 5% of antigens on RBCs for invasion, so individuals lacking Duffy
the total RBCs parasitized, is usually present in severe malaria.45 antigens are resistant to infection with P. vivax. The expansion
Major complications of severe malaria include respiratory dis- of the Duffy-negative population in West Africa seems to be an
tress syndrome, metabolic acidosis, circulatory shock, renal effective genetic adaptation, because P. vivax infection is almost
failure, hepatic failure, hypoglycemia, severe anemia (defined nonexistent in West Africa.40
as a hemoglobin level below 5g/dL),45 poor pregnancy Polymorphisms in genes for the and hemoglobin chains
outcome, and cerebral malaria.40 Even with treatment, the (Hb S, Hb C, Hb E, and - and -thalassemia) and for glucose-
fatality rate of severe malaria is 10% to 20%.45 6-phosphate dehydrogenase (G6PD) protect individuals from
The causes of anemia in malaria include (1) direct lysis of developing severe falciparum malaria. Although individuals
infected RBCs during schizogony, (2) immune destruction of with these polymorphisms become infected, they do not
infected and noninfected RBCs in the spleen, and (3) inhibi- develop serious complications.41 The reason for this protection
tion of erythropoiesis and ineffective erythropoiesis.42,44 The has not been completely elucidated. Enhanced phagocytosis of
destruction of noninfected RBCs contributes significantly to ring forms (sickle cell trait, -thalassemia trait, and G6PD defi-
the anemia. In the invasion process, parasites shed proteins ciency),51 decreased expression of PfEMP1 on infected erythro-
that bind to infected as well as noninfected RBCs.46 These cytes causing reduced endothelial adherence (sickle cell trait,
proteins may change the RBC membrane in noninfected cells, Hb C disease and trait),52,53 and decreased RBC invasion (Hb
allowing adherence of immunoglobulins and complement and E trait)54 may contribute to this protective effect. Over thou-
thus enhancing their removal by the spleen.46,47 In addition, sands of years, the evolutionary pressure of P. falciparum has
parasite proteins may also cause oxidative damage resulting in resulted in a higher frequency of these polymorphisms in pop-
decreased RBC survival.48 ulations located in high-prevalence malaria regions, including
Malaria parasites metabolize hemoglobin, forming toxic sub-Saharan Africa, the Middle East, and Southeast Asia (see
hemozoin or malaria pigment. When RBCs lyze in schizogony, Figure 26-3). Polymorphisms in cytokines and cellular recep-
the hemozoin and other toxic metabolites are released, which tors are also being investigated as modulators of malaria
results in an inflammatory response and cytokine imbalance.46 severity.40
Abnormal levels of tumor necrosis factor- and interferon-
result in inhibition of erythropoiesis as well as ineffective Clinical and Laboratory Findings
erythropoiesis; increased levels of interleukin-6 stimulate hep- The clinical symptoms of malaria are variable and can include
cidin production in the liver, which decreases the iron available fever, chills, rigors, sweating, headache, muscle pain, nausea,
to developing RBCs (see Chapter 19).46 In areas where malaria and diarrhea. In severe malaria, jaundice, splenomegaly, hepa-
is endemic, poor nutrition and coinfection with hookworm or tomegaly, shock, prostration, bleeding, seizures, or coma may
HIV contribute to the anemia, inflammation, and cytokine occur.45 In patients with chronic malaria or with repeated
imbalance.44,46 malarial infections, the spleen may be massively enlarged.
P. falciparum is unique and particularly lethal in that infected During fevers, the WBC count is normal to slightly increased,
RBCs adhere to endothelial cells in the microvasculature of but neutropenia may develop during chills and rigors. In
internal organs, including the brain, heart, lung, liver, kidney, chronic malaria with anemia, the reticulocyte count is decreased
dermis, and placenta.40,41 This contributes to the pathogenesis due to the negative effect of the inflammation on erythropoi-
by (1) obstructing the microvasculature and decreasing oxygen esis. In severe malaria, one or more of the following laboratory
344 PART IV Hematopathology: Erythrocyte Disorders

BOX 24-3 Thick Film Preparation for Malaria


To make a thick film, place three small drops of blood close together
near one end of the slide. With one corner of a clean slide, stir the
blood for about 30 seconds to mix the three drops over an area
approximately 1 to 2cm in diameter. Allow the film to dry thoroughly.
Stain the film using a water-based Giemsa stain. (The water-based A
stain lyses the red blood cells. Thin blood films are fixed in methyl
B
alcohol to preserve the red blood cells so they do not lyse.) In a thick
film, more parasites are seen in each field.

features are found: metabolic acidosis, decreased serum glucose


(less than 40mg/dL), increased serum lactate, increased serum
creatinine, decreased hemoglobin (less than 5g/dL), hemoglo- Figure 24-5 Peripheral blood film with a platelet on top of a red blood
binuria, and hyperparasitemia.45 cell (A) compared with an intraerythrocytic Plasmodium vivax ring form (B)
(1000).
Microscopic Examination
Malarial infection can be diagnosed microscopically by dem-
onstration of the parasites in the peripheral blood. Optimally,
blood should be collected before treatment is initiated.42,55 At
least two thick and two thin peripheral blood films should be
made as soon as possible after collection of venous blood in
ethylenediaminetetraacetic acid (EDTA) anticoagulant.55 Alter-
natively, blood films can be made directly from a capillary
puncture. Wright-Giemsa stain is used for visualization of the
parasites.42 Thick blood films concentrate the parasites and are
ideal for initial screening of peripheral blood. They are stained
with a water-based Wright-Giemsa stain without methanol
fixation to lyse the RBCs (Box 24-3). Thin blood films are used
for species identification and determination of the percent
parasitemia; they are stained after methanol fixation. The
percent parasitemia is determined by counting the number of
Figure 24-6 Two Plasmodium vivax trophozoites in a thin peripheral
parasitized RBCs (asexual stages) among 500 to 2000 RBCs on
blood film. Note that the infected red blood cells are enlarged and contain
a thin peripheral blood film and converting to a percentage.55 Schffner stippling, and the trophozoites are large and ameboid in
At least 300 fields on the thick and thin blood films should be appearance.
examined with the 100 objective before a negative result is
reported.42,55 Multiple samples taken at 8- to 12-hour intervals
may be needed, because the number of circulating parasites except the early ring forms. The growing trophozoite is ameboid
may vary with the timing of the erythrocytic schizogony.55 in appearance and has fine light brown hemozoin pigment.
Microscopy can detect 5 to 20 parasites per microliter of blood, The mature trophozoite almost fills the RBC (Figure 24-6). In
or 0.0001% parasitemia.42,56 A negative result for a single set an immature schizont, red chromatin begins to divide into two
of thick and thin peripheral blood films does not rule out a or more dots, and the mature schizont has 12 to 24 merozoites
diagnosis of malaria.42 Malarial parasite detection and species (Figure 24-7). Gametocytes are round with blue cytoplasm and
identification require experienced laboratory personnel. A light brown pigment; they have either centrally located (micro-
platelet lying on top of an erythrocyte in a thin blood film may gametocyte) or eccentrically located (macrogametocyte) red
be confused with a malarial parasite by an inexperienced chromatin. The length of the erythrocytic cycle is 44 to 48
observer (Figure 24-5). hours.42 P. vivax is difficult to eradicate, because the hypnozoite
forms remain dormant in the liver and may cause a relapse.
Plasmodium vivax. P. vivax is widely distributed and
causes 40% of human malaria cases worldwide.45 It is the pre- Plasmodium ovale. P. ovale is found mainly in West
dominant species in Asia and South and Central America, but Africa and India.57 The young ring forms are larger and more
also occurs in Southeast Asia, Oceania, and the Middle East.57 ameboid than those of P. vivax, and as the trophozoite grows,
It is rare in Africa and virtually absent in West Africa.57 The the RBCs become enlarged, oval, and fringed (Figure 24-8).
early ring forms are delicate with a red chromatin dot and Schffner stippling is present in all stages, including early ring
blue-staining cytoplasm. As the trophozoite grows, the RBC forms. In the schizont stage, the RBCs are oval, and the parasite
becomes enlarged. Schffner stippling appears in all stages is round and compact. The mature schizont has 8 to 12
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 345

Rights were not granted to include this figure Rights were not granted to include this figure
in electronic media. in electronic media.
Please refer to the printed publication. Please refer to the printed publication.

Figure 24-7 Plasmodium vivax schizont in a thin peripheral blood film. Figure 24-9 Plasmodium malariae band form in a thin peripheral blood
Note the number of merozoites and the presence of brown hemozoin pigment. film. (From Marler LM, etal: Parasitology image atlas, Indianapolis, 2003,
(From Marler LM, etal: Parasitology image atlas, Indianapolis, 2003, Indiana Indiana Pathology Images [CD-ROM].)
Pathology Images [CD-ROM].)

Rights were not granted to include this figure


Rights were not granted to include this figure in electronic media.
in electronic media. Please refer to the printed publication.
Please refer to the printed publication.

Figure 24-8 Two Plasmodium ovale trophozoites in a thin peripheral Figure 24-10 Plasmodium falciparum crescent-shaped gametocyte and
blood film. Note that the infected cells are enlarged, are oval, have fringed ring forms in a thin peripheral blood film (1000). (From Marler LM, etal:
edges, and contain Schffner stippling. (From Marler LM, etal: Parasitology Parasitology image atlas, Indianapolis, 2003, Indiana Pathology Images
image atlas, Indianapolis, 2003, Indiana Pathology Images [CD-ROM].) [CD-ROM].)

merozoites. Gametocytes are smaller than those of P. vivax, but Dominican Republic.57 It is also found in Asia, Southeast Asia,
have a similar appearance. The length of the erythrocytic cycle the Philippines, Indonesia, and South America.57 P. falciparum
is 48 hours.42 can produce high parasitemia (greater than 50% of RBCs
infected) due to its ability to invade RBCs of all ages. Only ring
Plasmodium malariae. P. malariae is found worldwide forms and crescent- or banana-shaped gametocytes are observed
but at low frequency. The highest prevalence is in East Africa in the peripheral blood. RBCs with trophozoites and schizonts
and India.57 The ring stage is often smaller and wider than that adhere to the endothelial cells in various organs and do not
of P. vivax, although the two may be indistinguishable. In circulate. Ring forms are small and can be easily missed. One
growing trophozoites, the cytoplasm forms a characteristic or more rings may occupy a given cell, with some rings located
narrow band across the cell and contains dark brown pigment at the cell periphery (see Figure 24-1). The crescent-shaped
(Figure 24-9). RBCs do not become enlarged, and there is no gametocytes have deep blue cytoplasm with brownish pigment
stippling. The mature schizont has 6 to 12 merozoites and and red chromatin near the center, and are easily recognized
often forms a rosette around clumped pigment. Gametocytes on blood films (Figure 24-10). The length of the erythrocytic
are similar to those of P. vivax, but are smaller. The length of cycle is 36 to 48 hours.42
the erythrocytic cycle is 72 hours.42
Plasmodium knowlesi. P. knowlesi is widespread in
Plasmodium falciparum. P. falciparum is the predomi- Malaysia, and cases have been reported in Myanmar, the
nant species in sub-Saharan Africa, Saudi Arabia, Haiti, and the Philippines, and Thailand.57 In the early trophozoite stage, it
346 PART IV Hematopathology: Erythrocyte Disorders

A B
Figure 24-11 Plasmodium knowlesi ring forms and trophozoite in a thin peripheral blood film (1000). (Courtesy Wadsworth Center Laboratories,
New York State Department of Health, New York, N.Y.)

can look similar to P. falciparum with high parasitemia and P. falciparum P. vivax P. malariae P. ovale
multiple ring forms in one cell. In the growing trophozoite,
the cytoplasm forms a band across the cell, similar to P. malar-
iae (Figure 24-11). Mature schizonts have an average of 16
merozoites and do not form rosettes.42 The length of the eryth-
rocytic cycle is only 24 hours, so infected patients can rapidly
develop a high level of parasitemia and severe malaria, and
prompt diagnosis and treatment are critical.34 Molecular
methods may be required for definitive diagnosis.
Figure 24-12 illustrates four species of Plasmodium.

Other Tests for Diagnosis


Fluorescent dyes may be use to stain Plasmodium species; they
are sensitive for parasite detection, but are not useful for specia-
tion.42 Molecular-based tests can be used for detection and
speciation of malarial parasites and are especially helpful in
cases of mixed infections and low parasitemia, as well as in the
identification of P. knowlesi. A dipstick-type rapid antigen test
has been approved by the Food and Drug Administration for
use in the United States. It is based on the detection of P. fal-
ciparum histidine-rich protein 2 and generic Plasmodium aldol-
ase.56 The overall sensitivity of the test is 95% for P. falciparum
and 69% for P. vivax, but the sensitivity decreases to 54% and
6%, respectively, when there are fewer than 100 parasites per
microliter of blood.42,56 Therefore, negative antigen test results
should be confirmed by microscopy.42,56

Treatment
Chloroquine or hydroxychloroquine is used for treatment of
all malaria, except for disease caused by strains of P. falciparum
and P. vivax acquired from areas known to harbor chloroquine-
resistant organisms.58 For infection with the latter strains, com- Figure 24-12 Malarial parasites from four species of Plasmodium. (From
bination therapy (use of two drugs with different mechanisms Diggs LW, Sturm D, Bell A: Morphology of human blood cells, ed 5, Abbott
Park, Ill, 1985, Abbott Laboratories. Permission has been granted with
of action) is recommended, including artemether-lumefantrine,
approval of Abbott Laboratories, all rights reserved by Abbott Laboratories,
atovaquone-proguanil, or quinine sulfate plus doxycycline or
Inc.)
tetracycline.45,58 Primaquine is also administered in P. vivax and
P. ovale infections to eradicate the hypnozoites in the liver and
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 347

prevent relapse.45,58 Prior to administration of primaquine,


testing for G6PD deficiency is recommended, because patients
with moderate to severe deficiency can develop hemolytic
anemia after primaquine treatment.45 There is growing concern
about the widespread resistance of P. falciparum to chloro-
quine, the emergence of chloroquine-resistant P. vivax strains,
and the possible emergence and spread of resistance to other
antimalarial drugs.45 Transfusion therapy is used for severe
anemia, including exchange transfusion for patients with a
level of parasitemia greater than 10% infected RBCs.58 Because
infections with P. falciparum or P. knowlesi have the potential
for a rapid, fatal course, patients must be treated without delay
after confirmation of the diagnosis. Intensive research is under-
way to develop a suitable vaccine.

Babesiosis Figure 24-13 Babesia microti ring forms in a peripheral blood film. Note
Babesiosis is a tick-transmitted disease caused by intraerythro- the varying appearance of the ring forms and the presence of multiple ring
cytic protozoan parasites of the genus Babesia. There are hun- forms in individual red blood cells (1000).
dreds of species of Babesia, but only a few are known to cause
disease in humans. B. microti is the most common cause of
babesiosis in the United States, where it was originally called congestive heart failure, renal shutdown, liver failure, or DIC.
Nantucket fever because the first cluster of cases was found on Severe disease may occur at any age, but is more common
Nantucket Island, off Massachusetts, in 1969 and the early in individuals over 50 years of age.60 Immune deficiency
1970s.59,60 The sexual cycle of B. microti occurs in the tick, Ixodes due to asplenia, malignancy, immunosuppressive drugs, or
scapularis, whereas its asexual cycle primarily occurs in the HIV infection increases the risk of severe disease.60,61 The
white-footed mouse, the reservoir host.61 Humans are inciden- overall mortality rate is less than 10%, but is likely higher in
tal hosts and become infected after injection of sporozoites immunocompromised individuals.60,66
during a blood meal by infected ticks. Other Babesia species are
found sporadically in humans and include B. duncani, B. diver- Laboratory Findings and Diagnosis
gens, B. divergens-like organisms, and B. venatorum.60 Babesia Evidence of hemolytic anemia is usually present in symptom-
may also be transmitted by transfusion of RBCs from asymp- atic infection, including decreased hemoglobin, increased
tomatic donors. Seventy cases of transfusion-transmitted babe- reticulocyte count, decreased serum haptoglobin level, and
siosis were reported between 1997 and 2007, and nine patients bilirubinemia. Leukopenia, thrombocytopenia, hemoglobin-
died.62 Only a few cases of possible congenitally acquired babe- uria, and proteinuria may also be present, along with abnormal
siosis have been reported.63 results on renal and liver function tests.60
The diagnosis of babesiosis is made by demonstration of
Geographic Distribution the parasite on Wright-Giemsastained thin peripheral blood
The areas in which B. microti is endemic in the United States films. Babesia appear as tiny rings or occasionally as tetrads
are southern New England, New York state, New Jersey, inside the RBCs. The ring forms may be round, oval, or
Wisconsin, and Minnesota.60,61 B. duncani occurs in northern ameboid; they have a dark purple chromatin dot and a minimal
California and Washington state; B. divergens-like organisms amount of blue cytoplasm surrounding a vacuole. Multiple
are found in Missouri, Kentucky, and Washington state; and B. rings can be found in one RBC (Figures 24-13 and 24-14).
divergens and B. venatorum occur in Europe.60,64 Isolated cases Tetrads may also appear in a Maltese cross formation. Babesia
of babesiosis have also been reported in Asia, Africa, and South can be distinguished from P. falciparum by the pleomorphism
America.60 of their ring forms, absence of hemozoin pigment and game-
tocytes, and occurrence of extraerythrocytic forms. Parasitemia
Clinical Findings may be low (fewer than 1% of RBCs affected) in early infec-
The incubation period for B. microti infection can range from tions, but as many as 80% of RBCs may be infected in asplenic
1 to 9 weeks.60 Infection is asymptomatic in perhaps a third of patients.67 In cases of low parasitemia, babesiosis can be diag-
individuals, so the exact prevalence is unknown.60,65 In other nosed by detection of IgG and IgM antibodies to B. microti by
individuals, B. microti causes a mild to severe hemolytic indirect immunofluorescent antibody assay.61 The sensitivity of
anemia.60,61 The patient usually experiences fever and nonres- the assay ranges from 88% to 96%.68 Definitive species identi-
piratory flulike symptoms, including chills, headache, sweats, fication requires PCR-based methods.60,61
nausea, arthralgias, myalgia, anorexia, and fatigue, that last
from several weeks to months. Jaundice, splenomegaly, or Treatment
hepatomegaly may be present. Some individuals progress to Babesiosis is treated with combination therapy using
severe, life-threatening disease due to acute respiratory failure, azithromycin/atovaquone or clindamycin/quinine.60 Exchange
348 PART IV Hematopathology: Erythrocyte Disorders

lymphadenopathy.74-76 Over several days, there is rapid hemo-


lysis, with the hematocrit dropping below 20% in two thirds of
patients.74 Reticulocytosis and mild leukocytosis with a left
shift accompany the anemia. The mortality rate is approxi-
mately 10% for hospitalized patients, and 90% for those who
are untreated.74-76 Diagnosis is made by blood culture and
observation of intraerythrocytic bacilli or coccobacilli on a
Wright-Giemsastained peripheral blood film. During the
acute phase, 2% to 100% of the RBCs can be infected.76
Serologic diagnosis with indirect immunofluorescent anti-
body or immunoblotting has a sensitivity of 89%.75 The
acute stage is treated with transfusions and ciprofloxacin or
chloramphenicol.75
Carrin disease was named for a Peruvian medical student,
Daniel Alcides Carrin, who in 1885 inoculated himself with
fluid from a wart of a patient in the chronic stage of infection.
Figure 24-14 Babesia microti ring and tetrad forms in a peripheral blood
film. (1000). He subsequently developed fatal hemolytic anemia similar to
that in Oroya fever, which linked the two stages of the disease
to the same agent.77

transfusion is used when greater than 10% of RBCs are infected


HEMOLYTIC ANEMIA CAUSED BY OTHER
or when there is evidence of major organ failure.60 Asymptom-
RED BLOOD CELL INJURY
atic infections usually require no treatment.60,61
Drugs and Chemicals
Clostridial Sepsis Hemolytic anemia of varying severity may result from drugs or
Sepsis with massive intravascular hemolysis, which is often chemicals that cause the oxidative denaturation of hemoglo-
fatal, is a rare complication of infection with Clostridium per- bin, leading to the formation of methemoglobin and Heinz
fringens, an anaerobic gram-positive bacillus. C. perfringens bodies.78 Examples of agents that can cause hemolytic anemia
grows very rapidly (7-minute doubling time) and produces an in individuals with normal RBCs include dapsone, a drug used
-toxin with phospholipase C and sphingomyelinase activity to treat leprosy and dermatitis herpetiformis,1,79 and naphtha-
that hydrolyzes RBC membrane phospholipids.69,70 The RBCs lene, a chemical found in mothballs.80 Individuals deficient
become spherical and extremely susceptible to osmotic lysis, in G6PD or other RBC enzymes are particularly sensitive to
which results in massive hemolysis and dark red plasma and the effects of oxidative agents. For example, primaquine can
urine.69,71,72 The hematocrit may drop to below 10%.71,72 The cause hemolytic anemia in G6PD-deficient individuals45 (see
intravascular hemolysis can trigger DIC and renal failure. Sphe- Chapter 23).
rocytes, microspherocytes, and toxic changes to neutrophils The typical laboratory findings include a decrease in hemo-
can be observed on a peripheral blood film.69,71,72 Some condi- globin, reticulocytosis, increase in serum indirect bilirubin,
tions that increase the risk of clostridial sepsis and hemolytic and decrease in serum haptoglobin level. In severe drug- or
anemia include malignancies (genitourinary, gastrointestinal, chemical-induced hemolytic anemia, Heinz bodies (denatured
and hematologic), solid organ transplant, postpartum or post- hemoglobin) may be observed in RBCs. Heinz bodies can only
abortion infections, and deep wounds.69,73 Swift therapy with be visualized with a supravital stain, and they appear as round,
transfusions, antibiotics, and fluid management is required. blue granules attached to the inner layer of the RBC membrane
The prognosis is grave, and many patients die despite intensive (see Figure 14-11). Exposure to high levels of arsine gas,
treatment. copper, and lead can also cause hemolysis.78

Bartonellosis Venoms
Human bartonellosis (Carrin disease) is transmitted by the Envenomation from contact with snakes, spiders, bees, or
bite of a female sandfly and is endemic in certain regions of wasps can induce hemolytic anemia in some individuals. The
Peru, Ecuador, and Columbia.74,75 It is caused by Bartonella hemolysis can occur acutely or be delayed one or more days
bacilliformis, a small, pleomorphic, intracellular coccobacillus after a bite or sting.78 The severity of the hemolysis depends on
that infects RBCs. There are two clinical stages: the first stage is the amount of venom injected, and in severe cases renal failure
characterized by acute hemolytic anemia (called Oroya fever and death can result. Some mechanisms by which venoms can
after a city in the Peruvian Andes); the second or chronic induce hemolysis are direct disruption of the RBC membrane,
verruga stage is characterized by the eruption of skin lesions alteration of the RBC membrane that results in complement-
and warts on the extremities, face, and trunk. The acute mediated lysis, and initiation of DIC.78 Hemolytic anemia has
phase begins with fever, malaise, headache, and chills, followed been reported after bites from poisonous snakes (e.g., some
by pallor, jaundice, hepatosplenomegaly, and general cobras and pit vipers) and the brown recluse spider (Loxosceles
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 349

reclusa), and after multiple stings (50 or more) by bees or


wasps.78,81-84

Extensive Burns (Thermal Injury)


Warming normal RBCs to 49 C in vitro induces RBC fragmen-
tation and budding.1 Likewise, patients with extensive burns
manifest similar RBC injury with acute hemolytic anemia.
Schistocytes, spherocytes, and microspherocytes are observed
on the peripheral blood film (Figure 24-15), but the damaged
RBCs are usually cleared by the spleen within 24 hours of the
burn injury.85 In addition to resulting from the acute hemoly-
sis, the anemia associated with extensive burns is also caused
by blood loss during surgical excision and grafting of the burn
wounds, nutritional deficiency, impaired metabolism, and
Figure 24-15 Peripheral blood film for a patient with extensive burns. anemia of chronic inflammation.86 Overheating blood in mal-
Note the presence of schistocytes, microspherocytes, and spherocytes functioning blood warmers prior to transfusion can also result
(1000).
in RBC fragmentation and hemolysis of the donor RBCs.1

SUMMARY
A common feature of the nonimmune extrinsic hemolytic anemias abrupt increase in blood pressure that damages the endothelial
is the presence of a condition that causes physical or mechanical cells and results in activation of coagulation factors and platelets;
injury to the RBCs. These conditions include microangiopathic about one fourth of patients have microangiopathic hemolytic
hemolytic anemia; macroangiopathic hemolytic anemia; some anemia.
infections; exposure to certain drugs, chemicals, or venoms; and DIC is due to the widespread intravascular activation of the
extensive burns. hemostatic system and formation of fibrin thrombi; coagulation
Microangiopathic hemolytic anemia is characterized by the shear factors and platelets are consumed in the thrombi and secondary
ing of RBCs as they pass through small blood vessels partially fibrinolysis occurs. Schistocytes are found in about half of
blocked by microthrombi. Fragmented RBCs (called schistocytes) the cases.
are formed, and the premature RBC destruction results in hemo Macroangiopathic hemolytic anemia is caused by traumatic
lytic anemia. Ischemic injury to the brain, kidney, and other organs cardiac hemolysis (RBC fragmentation from damaged or prosthetic
also occurs. Microangiopathic hemolytic anemia can occur in TTP, cardiac valves) or exercise-induced hemolysis (mechanical trauma
HUS, HELLP syndrome, DIC, and hypertensive crisis. from forceful impact on feet or hands or strenuous exercise). The
TTP is a rare disorder found predominantly in adults and charac platelet count is normal in both conditions; schistocytes are seen
terized by microangiopathic hemolytic anemia, severe throm only in traumatic cardiac hemolysis.
bocytopenia, increased level of lactate dehydrogenase, and Infections associated with hemolytic anemia due to invasion of
variable symptoms of fever, neurologic dysfunction, and renal RBCs include malaria, babesiosis, and bartonellosis. Hemolysis in
failure. Acquired TTP is due to autoimmune inhibition of the clostridial sepsis is due to the production of -toxin.
VWF-cleaving protease ADAMTS 13; it also occurs secondary to In malaria, severe anemia is due to direct lysis of infected RBCs,
other conditions such as stem cell transplantation, disseminated immune destruction of infected and uninfected RBCs in the spleen,
cancer, pregnancy, and use of certain drugs, as well as and inhibition of erythropoiesis.
from unknown causes. Familial TTP is due to mutations in the Plasmodium (five species) and Babesia organisms are identified
ADAMTS13 gene. by the morphology of their intraerythrocytic stages on a Wright-
HUS is characterized by microangiopathic hemolytic anemia, Giemsastained peripheral blood film. Plasmodia are transmitted
thrombocytopenia, and acute renal failure. D+ HUS (90% of cases) to humans by mosquitoes, whereas a tick is the vector for babesia.
is found predominantly in young children and is caused by toxin- Hemolytic anemia can also be caused by injury to RBCs by drugs,
producing strains of E. coli. Atypical HUS is due to mutations in chemicals, venoms, and extensive burns (thermal injury). In
genes coding for complement components. It can also occur sec patients with extensive burns, schistocytes, spherocytes, and
ondary to organ transplantation, cancer, pregnancy, HIV infection, microspherocytes are observed on the peripheral blood film.
and some drugs.
HELLP syndrome is a serious complication of pregnancy present Now that you have completed this chapter, go back and
ing with microangiopathic hemolytic anemia, thrombocytopenia, read again the case study at the beginning and respond
and elevated levels of liver enzymes. Hypertensive crises is an to the questions presented.
350 PART IV Hematopathology: Erythrocyte Disorders

R E V I E W Q UESTIONS
1. Which one of the following is a feature found in all micro- c. Competition for vitamin B12 in the erythrocyte
angiopathic hemolytic anemias? d. Immune destruction of noninfected RBCs in the spleen
a. Pancytopenia
b. Thrombocytosis 8. Which Plasmodium species is widespread in Malaysia, has
c. Intravascular RBC fragmentation RBCs with multiple ring forms, has band-shaped early
d. Prolonged prothrombin time and partial trophozoites, shows a 24-hour erythrocytic cycle, and can
thromboplastin time cause severe disease and high parasitemia?
a. P. falciparum
2. Typical laboratory findings in TTP and HUS include: b. P. vivax
a. Schistocytosis and thrombocytopenia c. P. knowlesi
b. Anemia and reticulocytopenia d. P. malariae
c. Increased levels of lactate dehydrogenase and
aspartate aminotransferase 9. One week after returning from a vacation in Rhode Island,
d. Increased levels of free plasma hemoglobin and a 60-year-old man experienced fever, chills, nausea, muscle
serum haptoglobin aches, and fatigue of 2 days duration. A complete blood
count (CBC) showed a WBC count of 4.5 109/L, hemo-
3. The pathophysiology of acquired TTP involves: globin of 10.5g/dL, a platelet count of 134 109/L, and
a. Shiga toxin damage to endothelial cells and a reticulocyte count of 2.7%. The medical laboratory sci-
obstruction of small blood vessels in glomeruli entist noticed tiny ameboid ring forms in some of the
b. Formation of platelet-VWF thrombi due to RBCs, and some tetrad forms in others. These findings
autoantibody inhibition of ADAMTS 13 suggest:
c. Overactivation of the complement system and a. Bartonellosis
endothelial cell damage due to loss of regulatory b. Malaria
function c. Babesiosis
d. Activation of the coagulation and fibrinolytic systems d. Clostridial sepsis
with fibrin clots throughout the microvasculature
10. What RBC morphology is characteristically found within
4. Which of the following tests yields results that are abnor- the first 24 hours following an extensive burn?
mal in DIC, but are usually within the reference range or a. Macrocytosis and polychromasia
just slightly abnormal in TTP and HUS? b. Burr cells and crenated cells
a. Indirect serum bilirubin and serum haptoglobin c. Howell-Jolly bodies and bite cells
b. Prothrombin time and partial thromboplastin time d. Schistocytes and microspherocytes
c. Lactate dehydrogenase and aspartate aminotransferase
d. Serum creatinine and serum total protein 11. A 36-year-old woman was brought to the emergency
department by her husband because she had experienced
5. Which one of the following laboratory results may be seen a seizure. He reported that she had been well until that
in hypertensive crises, traumatic cardiac hemolytic anemia, morning, when she complained of a sudden headache and
and exercise-induced hemoglobinuria? malaise. She was not taking any medications and had no
a. Schistocytes on the peripheral blood film history of previous surgery or pregnancy. Laboratory
b. Thrombocytopenia studies showed a WBC count of 15 109/L, hemoglobin
c. Decreased serum haptoglobin of 7.8g/dL, a platelet count of 18 109/L, and schistocytes
d. Hemosiderinuria and helmet cells on the peripheral blood film. Chemistry
test results included a markedly elevated level of lactate
6. Which of the following species of Plasmodium produce dehydrogenase and a slight increase in the level of total
hypnozoites that can remain dormant in the liver and and indirect serum bilirubin. The urinalysis results were
cause a relapse months or years later? positive for protein and blood, but there were no RBCs in
a. P. falciparum the sediment. Prothrombin time and partial thromboplas-
b. P. vivax tin time were within the reference range. When the entire
c. P. knowlesi clinical and laboratory picture is considered, which of the
d. P. malariae following is the most likely diagnosis?
a. HUS
7. Which one of the following is not a mechanism causing b. HELLP syndrome
anemia in P. falciparum infections? c. TTP
a. Inhibition of erythropoiesis d. Hypertensive crisis
b. Lysis of infected RBCs during schizogony
CHAPTER 24 Extrinsic Defects Leading to Increased Erythrocyte DestructionNonimmune Causes 351

REFERENCES
1. Schrier SL, Price EA: Extrinsic nonimmune hemolytic anemias. 21. Marik PE, Varon J: Hypertensive crises: challenges and man-
In Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology: agement. Chest 131:1949-1962, 2007.
basic principles and practice, ed 5, Philadelphia, 2009, Churchill 22. van den Born BJH, Honnebier UPF, Koopmans RP, et al:
Livingstone, pp 659-667. Microangiopathic hemolysis and renal failure in malignant
2. Moake JL: Thrombotic microangiopathies. N Engl J Med hypertension. Hypertension 45:246-251, 2005.
347:589-600, 2002. 23. Garewal M, Yahya S, Ward C, et al: MRI changes in throm-
3. Moake J: Thrombotic microangiopathies: multimers, metal- botic microangiopathy secondary to malignant hypertension.
loprotease, and beyond. Clin Transl Sci 2:366-373, 2009. J Neuroimaging 17:178-180, 2007.
4. Yu M, Nardella A, Pechet L: Screening tests of disseminated 24. Shapira Y, Vaturi M, Sagie A: Hemolysis associated with pros-
intravascular coagulation: guidelines for rapid and specific thetic heart valves. a review. Cardiol Rev 17:121-124, 2009.
laboratory diagnosis. Crit Care Med 28:1777-1780, 2000. 25. Davidson RJL: Exertional haemoglobinuria: a report on three
5. Sadler JE: Thrombotic thrombocytopenic purpura: a moving cases with studies on the haemolytic mechanism. J Clin Pathol
target. Hematology Am Soc Hematol Educ Program, 2006, 17:536-540, 1964.
pp 415-420. 26. Telford RD, Sly GJ, Hahn AG, et al: Footstrike is the major
6. Zheng XL, Kaufman RM, Goodnough LT, et al: Effect of cause of hemolysis during running. J Appl Physiol 94:38-42,
plasma exchange on plasma ADAMTS13 metalloprotease 2003.
activity, inhibitor level, and clinical outcome in patients with 27. Selby GB, Eichner ER: Endurance swimming, intravascular
idiopathic and nonidiopathic thrombotic thrombocytopenic hemolysis, anemia, and iron depletion. New perspective on
purpura. Blood 103:4043-4049, 2004. athletes anemia. Am J Med 81:791-794, 1986.
7. Kremer Hovinga JA, Vesely SK, Terrell DR, et al: Survival and 28. Shaskey DJ, Green GA: Sports haematology. Sports Med 29:27-
relapse in patients with thrombotic thrombocytopenic 38, 2000.
purpura. Blood 115:1500-1511, 2010. 29. Tobal D, Olascoaga A, Moreira G, et al: Rust urine after
8. Moake JL, Rudy CK, Troll JH, et al: Unusually large plasma intense hand drumming is caused by extracorpuscular hemo-
factor VIII: von Willebrand factor multimers in chronic lysis. Clin J Am Soc Nephrol 3:1022-1027, 2008.
relapsing thrombotic thrombocytopenic purpura. N Engl J 30. Smith JA, Kolbuch-Braddon M, Gillam I, et al: Changes in the
Med 307:1432-1435, 1982. susceptibility of red blood cells to oxidative and osmotic
9. Sadler JE: Von Willebrand factor, ADAMTS13, and throm- stress following submaximal exercise. Eur J Appl Physiol
botic thrombocytopenic purpura. Blood 112:11-17, 2008. 70:427-436, 1995.
10. Furlan M, Robles R, Galbusera M, et al: Von Willebrand 31. Beneke R, Bihn D, Htler M, et al: Haemolysis caused by
factorcleaving protease in thrombotic thrombocytopenic alterations of - and -spectrin after 10 to 35min of severe
purpura and the hemolytic-uremic syndrome. N Engl J Med exercise. Eur J Appl Physiol 95:307-312, 2005.
339:1578-1584, 1998. 32. Yusof A, Leithauser RM, Roth HJ, et al: Exercise-induced
11. Tsai H-M, Lian E C-Y: Antibodies to von Willebrand factor hemolysis is caused by protein modification and most
cleaving protease in acute thrombotic thrombocytopenic evident during the early phase of an ultraendurance race.
purpura. N Engl J Med 339:1585-1594, 1998. J Appl Physiol 102:582-586, 2007.
12. Verbeke L, Delforge M, Dierickx D: Current insight into 33. Singh B, Lee KS, Matusop A, et al: A large focus of naturally
thrombotic thrombocytopenic purpura. Blood Coagul Fibrino- acquired Plasmodium knowlesi infections in human beings.
lysis 21:3-10, 2010. Lancet 363:1017-1024, 2004.
13. Scheiring J, Rosales A, Zimmerhackl LB: Clinical practice: 34. Cox-Singh J, Davis TME, Lee KS, et al: Plasmodium knowlesi
todays understanding of the haemolytic uraemic syndrome. malaria in humans is widely distributed and potentially life-
Eur J Pediatr 169:7-13, 2010. threatening. Clin Infect Dis 46:165-171, 2008.
14. Petruzziello TN, Mawji IA, Khan M, et al: Verotoxin biology: 35. Lee KS, Cox-Singh J, Brooke G, et al: Plasmodium knowlesi
molecular events in vascular endothelial injury. Kidney Int from archival blood films: further evidence that human infec-
75:S17-S19, 2009. tions are widely distributed and not newly emergent in
15. Delvaeye M, Noris M, De Vriese A, et al: Thrombomodulin Malaysian Borneo. Int J Parasitol 39:1125-1128, 2009.
mutations in atypical hemolytic-uremic syndrome. N Eng J 36. World Health Organization: World malaria report 2009.
Med 361:345-357, 2009. Geneva, 2009, WHO Press. Available at: http://www.who.int/
16. Norris M, Remuzzi G: Atypical hemolytic-uremic syndrome. malaria/world_malaria_report_2009/en/. Accessed April 25,
N Eng J Med 361:1676-1687, 2009. 2010.
17. Haram K, Svendsen E, Abildgaard U: The HELLP syndrome: 37. Centers for Disease Control and Prevention: Malaria facts.
clinical issues and management. BMC Pregnancy Childbirth Available at: http://www.cdc.gov/malaria/about/facts.html.
9:1-15, 2009. Accessed April 25, 2010.
18. Baker KR, Moake J: Hemolytic anemia resulting from physical 38. Centers for Disease Control: 2008 U.S. Malaria Data. Avail-
injury to red cells. In Kaushansky K, Lichtman MA, Beutler able at: http://www.cdc.gov/malaria/features/2008_us_data.
TJ, et al, editors: Williams hematology, ed 8, New York, html. Accessed April 25, 2010.
2010, McGraw-Hill Medical, chap 50. Available at: http:// 39. Mali S, Steele S, Slutsker L, et al: Malaria surveillanceUnited
www.accessmedicine.com/content.aspx?aID=6122396. States, 2007. MMWR Morb Mortal Wkly Rep 58:1-16, 2009.
Accessed April 3, 2010. 40. Miller LH, Baruch DI, Marsh K, et al: The pathogenic basis of
19. Mihu D, Costin N, Mihu CM, et al: HELLP syndromea malaria. Nature 415:673-679, 2002.
multisystemic disorder. J Gastrointest Liver Dis 16:419-424, 41. Wellems TE, Hayton K, Fairhurst RM: The impact of malaria
2007. parasitism: from corpuscles to communities. J Clin Invest
20. Levi M, Seligsohn U: Disseminated intravascular coagulation. 119:2496-2505, 2009.
In Kaushansky K, Lichtman MA, Beutler TJ, et al, editors: 42. Garcia LS: Malaria. Clin Lab Med 30:93-129, 2010.
Williams hematology, ed 8, New York, 2010, McGraw-Hill 43. Centers for Disease Control and Prevention: Local transmission
Medical, chap 130. Available at: http://www.accessmedicine. of Plasmodium vivax malariaPalm Beach County, Florida,
com/content.aspx?aID=6235198. Accessed July 4, 2010. 2003. MMWR Morb Mortal Wkly Rep 52:908-911, 2003.
352 PART IV Hematopathology: Erythrocyte Disorders

44. Ekvall H: Malaria and anemia. Curr Opin Hematol 10:108-114, humans and its differentiation from other piroplasms. Int J
2003. Parasitol 36:779-789, 2006.
45. World Health Organization: Guidelines for treatment of malaria, 65. Krause PJ, McKay K, Gadbaw J, et al: Increasing health burden
ed 2, Geneva, 2010, WHO Press. of human babesiosis in endemic sites. Am J Trop Med Hyg
46. Haldar K, Mohandas N: Malaria, erythrocytic infection, 68:431-436, 2003.
and anemia. Hematology Am Soc Hematol Educ Program, 2009, 66. Meldrum SC, Birkhead GS, White DJ, et al: Human babesiosis
pp 87-93. in New York State: an epidemiological description of 136
47. Waitumbi JN, Opollo MO, Muga RO, et al: Red cell surface cases. Clin Infect Dis 15:1019-1023, 1992.
changes and erythrophagocytosis in children with severe Plas- 67. Blevins SM, Greenfield RA, Bronze MS: Blood smear analysis
modium falciparum anemia. Blood 95:1481-1486, 2000. in babesiosis, ehrlichiosis, relapsing fever, malaria, and
48. Griffiths MJ, Ndungu F, Baird KL, et al: Oxidative stress and Chagas disease. Cleve Clin J Med 75:521-530, 2008.
erythrocyte damage in Kenyan children with severe Plasmo- 68. Krause PJ, Telford SR 3rd, Ryan R, et al: Diagnosis of babesio-
dium falciparum malaria. Br J Haematol 113:486-491, 2001. sis: evaluation of a serologic test for the detection of B. microti
49. Craig A, Scherf A: Molecules on the surface of the Plasmodium antibody. J Infect Dis 169:923-926, 1994.
falciparum infected erythrocyte and their role in malaria 69. Kapoor JR, Montiero B, Tanoue L, et al: Massive intravascular
pathogenesis and immune evasion. Mol Biochem Parasitol hemolysis and a rapid fatal outcome. Chest 132:2016-2019,
115:129-143, 2001. 2007.
50. Bridges DJ, Bunn J, van Mourik JA, et al: Rapid activation of 70. Urbina P, Flores-Daz M, Alape-Girn A, et al: Phospholipase
endothelial cells enables Plasmodium falciparum adhesion to C and sphingomyelinase activities of the Clostridium perfrin-
platelet-decorated von Willebrand factor strings. Blood gens -toxin. Chem Phys Lipids 159:51-57, 2009.
115:1472-1474, 2010. 71. McArthur HL, Dalal BI, Kollmannsberger C: Intravascular
51. Ayi K, Turrini F, Piga A, et al: Enhanced phagocytosis of ring- hemolysis as a complication of Clostridium perfringens sepsis.
parasitized mutant erythrocytes: a common mechanism that J Clin Oncol 24:2387-2388, 2006.
may explain protection against falciparum malaria in sickle 72. Merino A, Pereira A, Castro P: Massive intravascular haemoly-
trait and beta-thalassemia trait. Blood 104:3364-3371, 2004. sis during Clostridium perfringens sepsis of hepatic origin. Eur
52. Fairhurst RM, Baruch DI, Brittain NJ, et al: Abnormal display J Haematol 84:278-279, 2009.
of PfEMP-1 on erythrocytes carrying haemoglobin C may 73. Barrett JP, Whiteside JL, Boardman LA: Fatal clostridial sepsis
protect against malaria. Nature 435:1117-1121, 2005. after spontaneous abortion. Obstet Gynecol 99:899-901, 2002.
53. Cholera R, Brittain NJ, Gillrie MR, et al: Impaired cytoadher- 74. Maguina C, Gotuzzo E: Bartonellosis new and old. Infect Dis
ence of Plasmodium falciparuminfected erythrocytes contain- Clin North Am 14:1-22, 2000.
ing sickle hemoglobin. Proc Natl Acad Sci U S A 105:991-996, 75. Huarcaya E, Maguina C, Torres R, et al: Bartonellosis
2008. (Carrions disease) in the pediatric population of Peru: an
54. Chotivanich K, Udomsangpetch R, Pattanapanyasat K, et al: overview and update. Braz J Infect Dis 8:331-339, 2004.
Hemoglobin E: a balanced polymorphism protective against 76. Maguina C, Garcia PJ, Gotuzzo E, et al: Bartonellosis (Carrions
high parasitemias and thus severe P. falciparum malaria. Blood disease) in the modern era. Clin Infect Dis 33:772-779, 2001.
100:1172-1176, 2002. 77. Garcia-Caceres U, Garcia FU: Bartonellosis. an immunode-
55. Centers for Disease Control and Prevention: Diagnostic pressive disease and the life of Daniel Alcides Carrion. Am J
proceduresblood specimens: specimen collection and Clin Pathol 95:S58-S66, 1991.
microscopic examination. Available at: http://www.dpd.cdc. 78. Means RT, Glader B: Acquired nonimmune hemolytic
gov/dpdx/HTML/DiagnosticProcedures.htm. Accessed June disorders. In Greer JP, Foerster J, Rodgers GM, et al, editors:
13, 2010. Wintrobes clinical hematology, ed 12, Philadelphia, 2009,
56. Murray CK, Gasser RA, Magill AJ, et al: Update on rapid Lippincott Williams & Wilkins, pp 1025-1028.
diagnostic testing for malaria. Clin Microbiol Rev 21:97-110, 79. Ranawaka RR, Mendis S, Weerakoon HS: Dapsone-induced
2008. haemolytic anemia, hepatitis and agranulocytosis in a leprosy
57. Centers for Disease Control and Prevention: Malaria map. patient with normal glucose-6-phosphate-dehydrogenase
Available at: http://cdc-malaria.ncsa.uiuc.edu/. Accessed June activity. Lepr Rev 79:436-440, 2008.
16, 2010. 80. Lim HC, Poulose V, Tan HH: Acute naphthalene poisoning
58. Centers for Disease Control and Prevention: Guidelines for following the non-accidental ingestion of mothballs. Singa-
treatment of malaria in the United States, May 18, 2009. pore Med J 50:e298-e301, 2009.
Available at: http://www.cdc.gov/malaria/pdf/treatmenttable. 81. Athappan G, Balaji MV, Navaneethan U, et al: Acute renal
pdf. Accessed June 15, 2010. failure in snake envenomation: a large prospective study.
59. Ruebush TK, II. Juranek DD, Spielman A, et al: Epidemiology Saudi J Kidney Dis Transpl 19:404-410, 2008.
of human babesiosis on Nantucket Island. Am J Trop Med Hyg 82. McDade J, Aygun B, Ware RE: Brown recluse spider (Loxosceles
30:937-941, 1981. reclusa) envenomation leading to acute hemolytic anemia in
60. Vannier E, Krause PJ: Update on babesiosis. Interdiscip Perspect six adolescents. J Pediatr 156:155-157, 2010.
Infect Dis 2009:984568, 2009. 83. Kolecki P: Delayed toxic reaction following massive bee
61. Centers for Disease Control and Prevention: Babesiosis. envenomation. Ann Emerg Med 33:114-116, 1999.
Available at: http://www.cdc.gov/babesiosis/. Accessed June 84. Paudel B, Paudel K: A study of wasp bites in a tertiary hospital
15, 2010. of western Nepal. Nepal Med Coll J 11:52-56, 2009.
62. Gubernot DM, Lucey CT, Lee KC, et al: Babesia infection 85. Bull BS, Herrmann PC: Hemolytic anemia resulting from
through blood transfusions: reports received by the US Food chemical and physical agents. In Kaushansky K, Lichtman
and Drug Administration, 1997-2007. Clin Infect Dis 48:25- MA, Beutler TJ, et al, editors, Williams hematology, ed 8, New
30, 2009. York, 2010, McGraw-Hill Medical, chap 51. Available at:
63. Sethi S, Alcid D, Kesarwala H, et al: Probable congenital http://www.accessmedicine.com/content.aspx?aID=6111475.
babesiosis in infant, New Jersey, USA. Emerg Infect Dis Accessed July 6, 2010.
15:788-791, 2009. 86. Posluszny JA, Gamelli RL: Anemia of thermal injury: com-
64. Conrad PA, Kjemtrup AM, Carreno RA, et al: Description of bined acute blood loss anemia and anemia of critical illness.
Babesia duncani n.sp. (Apicomplexa: Babesiidae) from J Burn Care Res 31:229-242, 2010.
Extrinsic Defects Leading to
Increased Erythrocyte Destruction
25
Immune Causes
Elaine M. Keohane

OUTLINE OBJECTIVES
Overview of Immune After completion of this chapter, the reader will be able to:
Hemolytic Anemias
Pathophysiology of 1. Define immune hemolytic anemia and indicate the 6. Describe three mechanisms of drug-induced immune
Immune Hemolysis types of antibodies involved. hemolysis.
Laboratory Findings in 2. Compare and contrast the mechanisms of immune 7. Compare and contrast the pathophysiology of
Immune Hemolytic hemolysis mediated by immunoglobulin M (IgM) and immune hemolysis due to drug-dependent and drug-
Anemia IgG antibodies. independent antibodies, including the related labora-
Autoimmune Hemolytic 3. Describe typical laboratory findings in immune tory findings.
Anemia hemolytic anemia and the importance of the direct 8. Describe two types of hemolytic transfusion reac-
Warm Autoimmune antiglobulin test (DAT). tions, the usual immunoglobulin class involved, the
Hemolytic Anemia 4. Compare and contrast four types of autoimmune typical site of hemolysis, and important laboratory
Cold Agglutinin Disease
hemolytic anemia in terms of the immunoglobulin findings.
Paroxysmal Cold
class involved, the temperature for optimal reactivity 9. Describe the cause, pathophysiology, and laboratory
Hemoglobinuria
Mixed-Type Autoimmune of the autoantibody, the proteins detected by the DAT findings in Rh and ABO hemolytic disease of the
Hemolytic Anemia on the patients red blood cells, the presence of fetus and newborn.
Drug-Induced Immune complement activation, the type and site of hemoly- 10. Given a patient history and results of a complete
Hemolytic Anemia sis, and the specificity of the autoantibody. blood count, peripheral blood film examination, per-
Mechanisms of 5. Relate results of the DAT to the pathophysiology tinent biochemical tests on serum and urine, and the
Drug-Induced Immune and clinical findings in autoimmune hemolytic direct and indirect antiglobulin tests, determine the
Hemolysis anemia. type of immune hemolysis.
Antibody Characteristics
Nonimmune Drug-
Induced Hemolysis
Treatment
Alloimmune Hemolytic
Anemias CASE STUDY
Hemolytic Transfusion
After studying the material in this chapter, the reader should be able to respond to the following
Reaction
case study:
Hemolytic Disease of the
Fetus and Newborn A 20-year-old woman sought medical attention for an elevated temperature and a possible insect
bite. Physical examination revealed a small bite wound on the right lateral area of the chest that
was tender and contained a clear fluid with no pus. Necrosis was not evident. A diffuse erythema-
tous rash also was observed on most of her body. The physician prescribed 40mL of liquid acet-
aminophen (Tylenol) and a 10-day regimen of cefuroxime (Ceftin). A secretion from the lesion
was subjected to culture, but there was no growth after 72 hours. Blood was drawn for cultures,
for which negative results were reported after 5 days. Four days later, the patient went to the emer-
gency department, complaining of being unable to walk. A CBC, urinalysis, urine and sputum
cultures, and chemistry profile were ordered.
Continued

353
354 PART IV Hematopathology: Erythrocyte Disorders

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

Patient Results Reference Range


9
WBCs (10 /L) 5.9 4.5-11.5
RBCs (1012/L) 1.40 4.00-5.40
Hb (g/dL) 4.2 12.0-15.0
Hct (%) 13 35-49

The platelet count was within the reference range. Sphe-


rocytes and polychromasia were observed on the peripheral
blood film (Figure 25-1). The film also revealed a slight
increase in neutrophilic bands and 2 nucleated RBCs per 100
WBCs (not shown in figure). An increase in urobilinogen
was noted on the routine urinalysis report, but the result for
blood was negative. Urine and sputum culture results were
negative. Significant findings from the chemistry profile Figure 25-1 Wright-stained peripheral blood film for the patient in the
included elevated levels of serum indirect bilirubin and case study (1000).
lactate dehydrogenase. Subsequently, a direct antiglobulin
test (DAT) and 3 units of red blood cells were ordered. A 2. What does the positive DAT result imply?
positive result on the DAT for IgG was reported. The indirect 3. What mechanisms can lead to the development of drug-
antiglobulin test on the patients serum and on an eluate induced immune hemolytic anemia?
prepared from the patients cells yielded negative results for 4. Describe the mechanism that is the most probable cause
all panel cells tested. of this patients anemia.
1. Is this type of anemia caused by an intrinsic or extrinsic 5. In addition to the transfusion, what additional action
defect? should be taken?

The degree of anemia varies from asymptomatic and mild to


OVERVIEW OF IMMUNE
severe and life-threatening.
HEMOLYTIC ANEMIAS
The immune hemolytic anemias may be classified into
Immune hemolytic anemia and nonimmune hemolytic anemia the following groups: autoimmune hemolytic anemia, drug-
are the two broad categories comprising the extrinsic hemolytic induced immune hemolytic anemia, and alloimmune hemo-
anemias, disorders in which red blood cells (RBCs) are struc- lytic anemia (Box 25-1).1,2 It is important to determine the
turally and functionally normal, but a condition outside of the cause of an immune hemolytic anemia so that the appropriate
RBCs causes premature hemolysis. The nonimmune extrinsic therapy can be administered to the patient.
hemolytic anemias are the result of physical or mechanical
injury to the RBCs and are covered in Chapter 24. The immune Pathophysiology of Immune Hemolysis
hemolytic anemias are conditions in which RBC survival is In immune hemolysis an antibody binds to an antigen on the
shortened due to an antibody-mediated mechanism. The surface of RBCs and that binding results in premature removal
antibody may be an autoantibody (directed against a self RBC of those cells from the circulation through extravascular or
antigen), an alloantibody (directed against an RBC antigen of intravascular hemolysis (see Chapter 22). The two classes or
another person), or an antibody directed against a drug taken isotypes of antibodies involved in most immune hemolytic
by the patient (or its metabolite). Some antibodies are able to anemias are immunoglobulin G (IgG) and IgM. IgG is a
activate the classical complement pathway, which results in the monomer in a Y-like structure with two identical heavy chains
attachment of activated complement proteins to the RBC mem- ( H chains) and two identical light chains (either or ) con-
brane. RBCs with bound antibody or complement may be nected by disulfide bonds.3 At the top of the Y-like structure
prematurely removed from the circulation extravascularly by are two antigen binding (Fab) domains, each formed from the
macrophages (due to their receptors for complement and the N-terminus of the variable domain of one light and one heavy
Fc component of antibody), intravascularly by complement- chain. IgG has one Fc domain (the stem of the Y) consisting
mediated hemolysis, by or a combination of both mecha- of the C-terminus of the two heavy chains. IgM is a pentamer
nisms.1 Anemia develops when the amount of hemolysis consisting of five monomeric units connected by disulfide link-
exceeds the ability of the bone marrow to replace the RBCs lost. ages at the C-termini of their heavy chains ( H chains).3,4
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 355

results in the binding of many C3b molecules on the RBC


BOX 25-1 Classification of Immune Hemolytic surface. C3b binds to C4bC2a to activate C5 to C5b. Next, C5b,
Anemias C6, C7, C8, and C9 form the membrane attack complex
Autoimmune hemolytic anemia (MAC). The MAC inserts into the lipid bilayer, forming a
Warm autoimmune hemolytic anemia (WAIHA) pore that allows water and small ions to enter the cell, which
Idiopathic eventually causes intravascular lysis.3 Negative regulators
Secondary inhibit various complement proteins and complexes in the
Lymphoproliferative disorders pathway to prevent uncontrolled activation and excessive
Nonlymphoid neoplasms hemolysis.1,4
Collagen-vascular disease Hemolysis mediated by IgM antibodies requires comple-
Immunodeficiency disorders ment and can result in both extravascular and intravascular
Viral infections hemolysis.5 When IgM molecules attach to the RBC surface in
Cold agglutinin disease (CAD) relatively low density, complement activation results in C3b
Idiopathic binding to the membrane, but complement inhibitors prevent
Secondary full activation of the pathway to the terminal membrane attack
Acute: infections (Mycoplasma pneumoniae, infectious complex.1,5 C3b-sensitized RBCs are destroyed by extravascular
mononucleosis, other viruses) hemolysis, predominantly by macrophages (Kupffer cells) in
Chronic: lymphoproliferative disorders the liver, which have C3b receptors. Some of the C3b on the
Paroxysmal cold hemoglobinuria (PCH) RBCs can be cleaved, however, which leaves the C3d fragment
Idiopathic on the cell. RBCs sensitized with only C3d are not prematurely
Secondary removed from circulation, because macrophages lack a C3d
Viral infections receptor.5 In severe cases of immune hemolysis involving
Syphilis heavy sensitization of RBCs with IgM antibody, significantly
Mixed-type autoimmune hemolytic anemia more complement is activated, which overwhelms the com
Drug-induced immune hemolytic anemia (DIIHA) plement inhibitors. In these cases, complement activation
Drug dependent proceeds from C1 to C9 and results in rapid intravascular
Drug independent hemolysis.5
Alloimmune hemolytic anemias Hemolysis mediated by IgG antibodies occurs with or
Hemolytic transfusion reaction (HTR) without complement and predominantly by extravascular
Hemolytic disease of the fetus and newborn (HDFN) mechanisms.5 RBCs sensitized with IgG are removed from cir-
culation by macrophages in the spleen, which have receptors
for the Fc component of IgG1 and IgG3.1,5 IgG antibodies are
Because the composition, structure, and size of IgG and IgM not efficient in activating complement; therefore, intravascular
are different, their properties and mechanisms in mediating hemolysis by full activation of complement from C1 to C9 is
hemolysis are also different. rare (except with anti-P in paroxysmal cold hemoglobinuria).5
The classical complement pathway is an important media- However, if there is a high density of IgG3 or IgG1 bound to
tor of immune hemolysis. The major proteins of the classical antigens on the RBCs, some complement is activated and C3b
complement pathway are designated C1 through C9, and their binds to the membrane. If both IgG and C3b are on the RBC
components or fragments are designated with lowercase suf- membrane, there is faster clearance from the circulation by
fixes. The first protein, C1, has three components: C1q, C1r, macrophages in both the spleen and liver.1,5 Often, IgG-
C1s. After an antibody binds antigen on the RBC surface, C1q sensitized RBCs are only partially phagocytized by macro-
must bind to two adjacent Fc domains to activate the pathway.4 phages, which results in the removal of some membrane.
Theoretically only one IgM molecule is needed for activation Spherocytes are the result of this process, and they are the
due to its pentameric structure with five Fc domains; however, characteristic cell of IgG-mediated hemolysis.5 The spherocytes
with the monomeric IgG, at least two molecules in close prox- are eventually removed from circulation by entrapment in the
imity are required.4 Therefore IgM antibodies are highly effec- red pulp of the spleen (splenic cords), where they are rapidly
tive in activating complement, whereas IgG antibodies are phagocytized by macrophages (see Chapter 8).5 The mecha-
unable to activate the pathway unless there is a sufficient nisms of immune hemolysis are summarized in Table 25-1.
number of IgG molecules on the RBC surface.4,5 In addition,
subclasses IgG3 and IgG1 have high binding affinity for C1q, Laboratory Findings in Immune
whereas subclasses IgG2 and IgG4 have minimal ability to Hemolytic Anemia
bind complement.4,5 Laboratory findings in immune hemolytic anemia are similar
The binding of C1q to adjacent Fc domains activates C1r, to the findings in other hemolytic anemias and include
which subsequently activates C1s. C1s activates C4 and then decreased hemoglobin (Hb), increased reticulocyte count,
C2, which results in the binding of a small number of C4bC2a increased levels of indirect serum bilirubin and lactate dehy-
complexes to the RBC membrane. The C4bC2a complex is an drogenase, and decreased serum haptoglobin level. If the
active C3 convertase enzyme that cleaves C3 in plasma; this hemolysis is predominantly intravascular, the haptoglobin
356 PART IV Hematopathology: Erythrocyte Disorders

TABLE 25-1 Major Mechanisms of Immune Hemolysis


IgM Mediated IgG Mediated
Extravascular IgM activation of classical complement pathway from C1 Clearance of IgG-sensitized RBCs by macrophages mainly in spleen
hemolysis to C3b only; clearance of C3b-sensitized RBCs by Formation of spherocytes by partial phagocytosis of IgG-sensitized RBCs;
macrophages mainly in liver spherocytes cleared by macrophages after entrapment in spleen
IgG* activation of classical complement pathway from C1 to C3b only;
requires high-density IgG on RBC surface; clearance of IgG- and
C3b-sensitized RBCs by macrophages in spleen and liver
Intravascular Full IgM activation of classical complement pathway from Full IgG activation of classical complement pathway from C1 to C9 and
hemolysis C1 to C9 and direct RBC lysis; requires high-density direct RBC lysis; requires very-high-density IgG on RBCs for activation
IgM on RBCs to overcome complement inhibitors and to overcome complement inhibitors; uncommon

Ig, Immunoglobulin; RBC, red blood cell.


*IgG3 and IgG1 are most efficient in complement activation.

level will be moderately to severely decreased, plasma hemo-


AUTOIMMUNE HEMOLYTIC ANEMIA
globin will be increased, and the patient may have hemoglo-
binuria or even hemosiderinuria (in cases of chronic hemolysis) Autoimmune hemolytic anemia (AIHA) is a rare disorder char-
(see Chapter 22). The mean cell volume may be increased due acterized by premature RBC destruction and anemia caused by
to the reticulocytosis, and leukocytosis and thrombocytosis autoantibodies that bind the RBC surface with or without
may be present due to the increased erythroid proliferation in complement activation. Autoantibodies may arise as a result of
the bone marrow.1,6 Findings on the peripheral blood film immune system dysregulation and loss of immune tolerance,
include polychromasia (due to the reticulocytosis), spherocytes exposure to an antigen similar to an autoantigen, B lymphocyte
(due to IgG-mediated membrane damage by macrophages), neoplasm, or other unknown reason.1,6 The type, amount, and
and RBC agglutination (in some cases of IgM autoantibodies).5 duration of antigen exposure and genetic and environmental
Nucleated RBCs, occasional schistocytes, and erythrophagocy- factors may also contribute to the development of autoanti-
tosis may also be observed on the peripheral blood film.1 bodies.5,6 The anemia can be mild or severe, and the onset
To determine if the hemolysis is due to an immune mecha- acute or gradual. The severity of the anemia depends on the
nism, a direct antiglobulin test (DAT) is performed. The DAT autoantibody characteristics (titer, ability to react at 37 C,
detects in vivo sensitization of the RBC surface by IgG, C3d, or ability to activate complement, and specificity and affinity for
both.2 In the DAT procedure, polyspecific antihuman globulin the autoantigen), antigen characteristics (density on RBCs,
(AHG) is added to saline-washed patient RBCs. Polyspecific immunogenicity), as well as patient factors (age; ability of the
AHG has specificity for the Fc portion of human IgG and bone marrow to compensate for the hemolysis; function of
complement component C3d and will agglutinate the RBCs if macrophages, complement proteins and regulators; and under-
a critical number of one or both molecules is present on the lying conditions).1,6
RBC surface.2 If the DAT result is positive with polyspecific The autoimmune hemolytic anemias may be divided into
AHG, then the cells are tested with monospecific anti-IgG and four major categories based on the characteristics of the auto-
anti-C3d to identify the type of sensitization. If IgG is detected antibody and the mechanism of hemolysis: warm autoim-
on the RBCs, elution procedures are used to remove the anti- mune hemolytic anemia, cold agglutinin disease, paroxysmal
body. The specificity of the antibody may be determined by cold hemoglobinuria, and mixed-type autoimmune hemolytic
assessing the reaction of the eluate with screening and panel anemia (Table 25-2).2,5,6
reagent RBCs (RBCs genotyped for the major RBC antigens)
using the indirect antiglobulin (AHG) test. Identification of any Warm Autoimmune Hemolytic Anemia
circulating alloantibodies or autoantibodies by the indirect Warm autoimmune hemolytic anemia (WAIHA) is the most
antiglobulin test is also important in the investigation.2 The commonly encountered autoimmune hemolytic anemia com-
DAT result may be negative in patients with some immune prising up to 70% of cases.5 The autoantibodies causing
hemolytic anemias.2 A positive DAT result has been reported WAIHA react optimally at 37 C, and the vast majority of them
in apparently healthy individuals and in blood donors with no are IgG.1 WAIHA is most commonly found in adults older than
evidence of hemolysis.7,8 In addition, other disorders beside 40 years of age and in children younger than 4 years of age.6
immune hemolytic anemia can cause a positive DAT finding.2 WAIHA may be classified as idiopathic or secondary. In patients
Therefore diagnosis of immune hemolytic anemia cannot rely with idiopathic WAIHA, the etiology is unknown. Secondary
solely on the DAT and must take into account the patient WAIHA may be found in many conditions such as lymphopro-
history, symptoms, recent medications, previous transfusions, liferative diseases (chronic lymphocytic leukemia, B-lymphocyte
coexisting conditions including pregnancy, as well as the lymphomas, Waldenstrm macroglobulinemia), nonlymphoid
results of the applicable hematologic, biochemical, and sero- neoplasms (thymoma and cancers of the colon, kidney,
logic tests.2,5 lung, and ovary), autoimmune disorders (rheumatoid arthritis,
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 357

TABLE 25-2 Characteristics of Autoimmune Hemolytic Anemias


Warm Autoimmune Cold Agglutinin Paroxysmal Cold Mixed-Type Autoimmune
Hemolytic Anemia Disease Hemoglobinuria Hemolytic Anemia
Immunoglobulin class IgG (rarely IgM, IgA) IgM IgG IgG, IgM
Optimum reactivity temperature 37 C 4 C; reactivity 4 C 4-37 C
of autoantibody extends to >30 C
Sensitization detected by direct IgG or IgG + C3d; only C3d C3d C3d IgG and C3d
antiglobulin test uncommon
Complement activation Variable Yes Yes Yes
Hemolysis Extravascular primarily Extravascular; rarely Intravascular Extravascular and intravascular
intravascular
Autoantibody specificity Panreactive or Rh complex; rarely I (most), i (some), P Panreactive; unclear specificity
specific Rh or other antigen Pr (rare)

Ig, Immunoglobulin.

scleroderma, polyarteritis nodosa, Sjgren syndrome, systemic clinically significant alloantibodies are also present. Donor cells
lupus erythematosus), immunodeficiency disorders, and viral that lack the antigens corresponding to all clinically significant
infections.1,6 alloantibodies identified are selected for transfusion.1,2,5
The onset of WAIHA is usually insidious with symptoms Anemia in WAIHA can be mild or severe, with RBC life span
of anemia (fatigue, dizziness, dyspnea), but some cases can sometimes reduced to 5 days or less.1 Laboratory findings
be acute and life-threatening with fever, jaundice, splenomeg- for serum and urine reflect the predominantly extravascular
aly, and hepatomegaly, especially in children with WAIHA hemolysis that occurs in IgG-mediated immune hemolysis.
secondary to viral infections.1,6 An underlying lymphopro Polychromasia and spherocytes are the typical findings on
liferative disorder is suggested in adults with massive spleno- the peripheral blood film (see Figure 25-1). Occasionally the
megaly, lymphadenopathy, fever, petechiae, ecchymosis, or WAIHA is accompanied by immune thrombocytopenic
renal failure.1,6 purpura and a decreased platelet count; that condition is
Although most autoantibodies that cause WAIHA are IgG, known as Evans syndrome.10
rare cases involving IgA autoantibodies as well as cases with In symptomatic but nonlife-threatening WAIHA, a cortico-
fatal outcomes caused by warm-reacting IgM antibodies have steroid such as prednisone is the initial treatment of choice.1,2
been reported.1,2,9 Hemolysis is predominantly extravascular Approximately 70% to 80% of patients show improvement
in WAIHA, and cases of fulminant intravascular hemolysis with prednisone, but most adult patients need to be on a long-
are rare.5 term maintenance dosage to remain asymptomatic.1,6 Children
The result of the DAT is positive in over 95% of patients, generally respond very well to corticosteroids and experience a
with approximately 85% of patients having IgG alone or both complete remission.6 Splenectomy is an option in patients
IgG and C3d on their RBCs, and 10% to 14% having C3d only. with chronic WAIHA that is refractive to prednisone therapy
Between 1% and 4% of patients have a negative DAT result.1,2,5 or requires long-term, high-dose prednisone therapy, and a
A negative DAT result may be caused by IgA or IgM autoanti- favorable response is achieved in 50% to 75% of patients.1,6
bodies that are not detected by the polyspecific AHG, by the Immunosuppressive drugs, such as cyclophosphamide or aza-
presence of IgG or C3d in an amount below the reagent detec- thioprine, are used for refractory WAIHA, but the side effects
tion limit, by the dissociation of IgG antibodies with low may be severe.1 Rituximab (monoclonal anti-CD20) has also
avidity during the washing phase of the DAT, or by various been used. It causes minimal side effects and produces a
technical errors.1,2 Therefore a negative DAT result does not rule response rate of 17% to 100% according to various published
out autoimmune hemolytic anemia. case reports.1 Intravenous gamma globulin or plasma exchange
Warm autoantibodies are usually panreactive; that is, they is also used in severe disease, but is not recommended for
will agglutinate all screening and panel cells, donor RBCs, and routine or long-term use.1,6 In secondary WAIHA, successful
the patients own RBCs, so the specificity of the autoantibody management of the underlying condition often controls the
is not apparent.2,5 In some cases Rh complex specificity can be hemolysis and anemia. Life-threatening WAIHA requires RBC
demonstrated.2 Rarely, a specific autoantibody to an antigen in transfusion. If the autoantibody has broad specificity, all RBC
the Rh blood group system is identified. Autoantibodies to units may be incompatible with the autoantibody.1,2 In such
other antigens (such as LW, Jk, K, Di, Ge, Lu, M, N, S, U, Ena, cases a minimum volume of RBCs is used, and the patient is
and Wrb) are rarely encountered.2,5,6 For most patients (approx- carefully monitored during the transfusion.
imately 80%) the autoantibody can be detected in the serum.
Because the autoantibody is panreactive, it may mask reactions Cold Agglutinin Disease
of alloantibodies with RBC panel cells. If an RBC transfusion Cold agglutinins are autoantibodies of the IgM class that react
is necessary, it is crucial to perform tests to determine if optimally at 4 C. Cold agglutinins are commonly found in
358 PART IV Hematopathology: Erythrocyte Disorders

healthy individuals. These nonpathologic cold agglutinins are Virtually all adult RBCs are positive for the I antigen, so anti-I
polyclonal, occur in low titers (less than 1:64 at 4 C), and will agglutinate all screening and panel cells, donor RBCs, and
have no reactivity above 30 C.1,5 Most pathologic cold agglu- the patients own RBCs at room temperature and higher,
tinins are monoclonal, occur at high titers (greater than 1:1000 depending on the thermal amplitude of the autoantibody.
at 4 C), and are capable of reacting at temperatures greater Anti-I will show weaker or negative reactions with cord RBCs
than 30 C.2,6 Because pathologic cold agglutinins can react at (cord cells are negative for I antigen, but positive for i antigen).2
body temperature, they may induce cold agglutinin disease A cold agglutinin method is used to determine the titer of
(CAD). Cold agglutinins that are able to bind RBC antigens the antibody at 4 C. Pathologic cold agglutinins can reach
near or at 37 C (high thermal amplitude) cause more severe titers of 1:10,000 to 1:1,000,000 at 4 C.1,5 Blood specimens
symptoms.1,11 CAD comprises approximately one fourth of the to determine the titer of cold agglutinins are maintained at 37
cases of autoimmune hemolytic anemia.6 C after collection to prevent the binding of the autoantibody
Chronic CAD is a rare hemolytic anemia that typically to the patients own RBCs, which can falsely decrease the anti-
occurs in middle-aged and elderly individuals.12 In one recent body titer in the serum. Alternatively, a sample anticoagulated
study, the median age at onset was 67 years, with a range of with ethylenediaminetetraacetic acid (EDTA) can be warmed
30 to 92 years.13 Chronic CAD can be idiopathic, with no for 10 to 15 minutes at 37 C to dissociate autoabsorbed anti-
known cause, or secondary due to lymphoproliferative neo- body prior to determining the titer.
plasms such as B-lymphocyte lymphomas, Waldenstrm mac- When a high-titer cold agglutinin is present, an EDTA-
roglobulinemia, or chronic lymphocytic leukemia.12 In chronic anticoagulated blood specimen can show visible agglutinates
CAD, the autoantibody is usually monoclonal IgM with light in the tube at room temperature or below.1 The agglutination
chains.2,5,11 can also be observed on a peripheral blood film (Figure 25-2).
Acute CAD occurs secondary to Mycoplasma pneumoniae Blood specimens from patients with cold agglutinins must be
infection, infectious mononucleosis, and other viral infections. warmed to 37 C for 15 minutes before analysis by automated
These cold agglutinins are polyclonal IgM with a normal dis- hematology analyzers. RBC agglutination grossly elevates the
tribution of and light chains.2,5 mean cell volume, reduces the RBC count, and has unpredict-
In CAD, the IgM autoantibody binds to RBCs after exposure able effects on other indices (see Chapter 39). When the sample
to the cold, particularly in the peripheral circulation and the is warmed at 37 C, the antibody dissociates from the RBCs,
vessels of the skin where temperatures can drop to 30 C.6,11 and agglutination usually disappears. If not, a new specimen
During the brief transit through these colder areas, IgM auto- is collected and maintained at 37 C for the entire time before
antibodies activate the classical complement pathway. When testing. To avoid agglutination on a peripheral blood film, the
the RBCs return to the central circulation, the IgM antibody slide can also be warmed to 37 C prior to the application of
dissociates, but C3b components remain on the cell.2,11 Hemo- blood. Cold agglutinins can also interfere with ABO typing.2
lysis is predominantly extravascular by hepatic macrophages, Acute CAD associated with infections is self-limited and
which have receptors for C3b.1,11 However, if the autoantibody the cold agglutinin titers are usually less than 1:4000.12 If
has a high thermal amplitude, or there is a deficiency in com- the hemolysis is mild, no treatment is required; however,
plement regulatory proteins, full complement activation and patients with severe hemolysis require transfusion and sup-
intravascular hemolysis can occur.1 portive care.12
In chronic CAD, the clinical manifestations are variable. Patients with chronic CAD and mild anemia are regularly
Most patients have a mild anemia with hemoglobin ranging monitored and advised to avoid cold temperatures.1 In chronic
from 9 to 12 g/dL,12 but others can develop life-threatening
anemia with hemoglobin levels falling below 5 g/dL, especially
after exposure to cold temperatures.5,13 Individuals often expe-
rience fluctuation between mild and severe symptoms, and
approximately half require transfusions over the course of the
disease.11 Symptoms include fatigue, weakness, dyspnea, pallor
(due to the anemia), and acrocyanosis.12 Acrocyanosis is a
bluish discoloration of the extremities (fingers, toes, feet, ear
lobes, nose) due to RBC autoagglutination, which causes local
capillary stasis.1,2,12 Some patients also have episodes of hemo-
globinuria, especially after exposure to cold temperatures.2,12
In contrast, patients with acute CAD may have mild to severe
hemolysis that appears abruptly within 2 to 3 weeks after the
onset of infectious mononucleosis, another viral infection, or
M. pneumoniae infection, but it resolves spontaneously within
days to a few weeks.12
The DAT result is positive because of the presence of C3d
on the RBC surface. The specificity of the cold agglutinin is Figure 25-2 Wright-stained peripheral blood film showing red blood cell
most often anti-I, but can be anti-i or, very rarely, anti-Pr.2 agglutination (500).
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 359

CAD with moderate to severe symptoms, rituximab (anti- intravascular hemolysis are found. Because the anti-P auto
CD20) produces partial remission in about half of patients, but antibody is dissociated from the RBCs at body temperature, the
median remission is less than a year.14,15 In a recent study, a DAT result is usually positive for C3d only.2
combination of rituximab with fludarabine resulted in a better The classic Donath-Landsteiner test for anti-P is done by
response rate (76%) and median duration of remission (66 collecting blood samples in two tubes, one for the patient test
months); however, hematologic toxicity was reported in almost and the other for the patient control.19 The patient test sample
half of the patients.16 Plasmapheresis may be used in severe is incubated first at 4 C for 30 minutes (to allow anti-P
cases but provides only temporary benefit.1 Corticosteroid binding to the P antigen and partial complement activation on
therapy and immunosuppressive therapy with cyclophospha- the RBCs), then at 37 C for 30 minutes (to allow full activa-
mide and chlorambucil are not effective for most patients.12 tion of the complement pathway to lysis). The patient control
Splenectomy is also not effective, because C3b-sensitized RBCs tube is kept at 37 C for both incubations (or a total of 60
in IgM-mediated autoimmune hemolysis are cleared primarily minutes). After centrifugation, the supernatant is examined for
by the liver.1,5,12 hemolysis. A positive test result for anti-P is indicated by hemo-
RBC transfusion is reserved for patients with life-threatening lysis in the patient test sample incubated first at 4 C and then
anemia or cardiovascular or cerebrovascular symptoms.1 If at 37 C, and no hemolysis in the patient control sample kept
transfusion is needed, the presence of clinically significant allo- at 37 C. In the control tube, the anti-P is not able to bind to
antibodies must be ruled out.2 In CAD cases involving an antigen at 37 C, so complement is not activated and hemo-
autoantibody with wide thermal amplitude, detection of coex- lysis does not occur. Initial test results may be falsely negative
isting warm-reactive alloantibodies can be time consuming due to low complement and/or anti-P levels in the patient
and difficult. During transfusion, the patient is kept warm, and sample because of the brisk hemolysis in vivo.19 Incubating
a blood warmer is used to minimize the in vivo reactivity of patient serum with complement and papain-treated compati-
the cold autoantibody.1 ble group O RBCs increases the sensitivity of the test in detect-
ing anti-P. The enzyme treatment provides greater exposure of
Paroxysmal Cold Hemoglobinuria the P antigen on the RBC surface for antibody binding.19
Paroxysmal cold hemoglobinuria (PCH) is an acute form of PCH is severe but self-limited and resolves in several days
cold-reactive hemolytic anemia. PCH can be idiopathic or sec- to a few weeks with an excellent prognosis.1,12 In most patients,
ondary. Historically, secondary PCH was associated with late- the anemia is severe and can be life-threatening, so transfusion
stage syphilis, but now it is most commonly seen in young is usually needed until the symptoms resolve. Because the
children after a respiratory viral infection.1,2,12 PCH is rare in anti-P autoantibody reacts only at lower temperatures and P
adults. The incidence of PCH in children has been reported to antigennegative blood is very rare, P-positive blood can be
be as high as 32% to 40% of children with autoimmune hemo- transfused.2
lytic anemia, with a median age at presentation of 5 years.17-19
The anti-P autoantibody, also called the Donath-Landsteiner Mixed-Type Autoimmune Hemolytic Anemia
antibody, is a complement-binding IgG hemolysin with speci- Mixed-type autoimmune hemolytic anemia occurs very infre-
ficity for the P antigen on RBCs. The anti-P autoantibody is quently.20 In this condition, the patient simultaneously devel-
biphasic in that at cold temperatures it binds to the P antigen ops an IgG autoantibody with optimum reactivity at 37 C
on RBCs and partially activates complement (C1 to C4), but (WAIHA) and a pathologic IgM autoantibody that reacts opti-
full complement activation (C3 through C9) and hemolysis mally at 0 C to 10 C but has a thermal amplitude of greater
occur only upon warming to 37 C.19 Exposure to cold tem- than 30 C (CAD).20,21 Patients with WAIHA and a nonpatho-
peratures is not required for the hemolytic manifestations in genic cold agglutinin (i.e., an agglutinin that does not react at
vivo, however, and the reasons for this have yet to be a temperature greater than 20 C) should not be classified as
explained.2,6 The anti-P autoantibody binds antigen optimally having a mixed-type autoimmune hemolytic anemia, because
at 4 C and has a thermal amplitude of less than 20 C. At the cold agglutinin is not clinically significant.12,20,21
warmer temperatures, the anti-P autoantibody dissociates from The hemolysis results from a combination of extravascular
the RBCs. The titer is usually less than 1:64.6,12 and intravascular mechanisms. The disease course appears to
Children typically present with acute fever, malaise, and be chronic with intermittent episodes of severe anemia.5,6,12,21
back, leg, and/or abdominal pain 1 to 2 weeks after an upper The DAT results can be positive with IgG only, C3d only, or
respiratory tract infection.6,12 Pallor, jaundice, and dark urine IgG and C3d.2 The warm autoantibody is typically panreactive
due to hemoglobinuria are frequently present.12 The abrupt with unclear specificity, whereas the cold-reacting antibody
onset of hemolysis causes a rapidly progressing and severe ane usually has anti-I specificity.5 Treatment is the same as that
mia, with hemoglobin levels often dropping below 5 gm/dL.12 described for WAIHA.5
Reticulocytosis is typical, but can be preceded by reticulo-
cytopenia.6,12 The peripheral blood film shows polychromasia
DRUG-INDUCED IMMUNE
and spherocytes, but schistocytes, nucleated RBCs, aniso
HEMOLYTIC ANEMIA
cytosis, poikilocytosis, and erythrophagocytosis can also be
observed.6,12 At first, leukopenia may be present; later, leukocy- Drug-induced immune hemolytic anemia (DIIHA) is very
tosis occurs.6,12 In addition, laboratory findings typical for rare, with an estimated annual incidence of about 1 per
360 PART IV Hematopathology: Erythrocyte Disorders

million persons.22 This condition is suspected when there is Drug-Dependent Antibodies


a sudden decrease in hemoglobin after administration of a Drug-dependent antibodies only react in vitro when the sus-
drug, clinical and biochemical evidence of extravascular or pected drug or its metabolite is present.2,24 There are two types
intravascular hemolysis, and a positive DAT result.23 Over 125 of drug-dependent antibodies:
drugs have been reported to cause DIIHA, with the most 1. Antibodies that react only with drug-treated cells2: These are IgG
common drug categories being antimicrobial, antiinflamma- drug antibodies that bind to the drug when it is strongly
tory, and antineoplastic drugs.24,25 The most common drugs associated with the RBC surface (drug adsorption mecha-
implicated in DIIHA in the last 10 years are cefotetan, ceftriax- nism). Because they have bound IgG, the RBCs are cleared
one, and piperacillin.22 Severe, even fatal, cases have been from the circulation extravascularly by macrophages in the
reported. spleen, and a hemolytic anemia gradually develops. Com-
plement is not usually activated. If the DIIHA is not recog-
Mechanisms of Drug-Induced nized, the patient may continue to take the drug to a point
Immune Hemolysis in which life-threatening anemia develops.2,22 Examples of
Various theories have been proposed to explain the mecha- drugs that elicit antibodies in this category are penicillin
nisms of DIIHA.22 Three generally accepted mechanisms and cefotetan.2,24 Laboratory features include a positive DAT
involve an antibody produced by the patient as a result of reaction with anti-IgG, whereas the reaction with anti-C3d
exposure to the drug and include (1) drug adsorption, (2) is usually negative. In the indirect AHG test, the patients
drugRBC membrane protein immunogenic complex, and (3) serum and an eluate of the patients cells react only with
RBC autoantibody induction. A fourth mechanism, drug- drug-treated RBCs and not with untreated RBCs.2,6
induced nonimmunologic protein adsorption, can result in a 2. Antibodies that react only in the presence of the drug2: These
positive DAT result, but no drug or RBC antibody is produced IgG and/or IgM antibodies bind to the drug or its metabo-
by the patient. This mechanism is discussed at the end of this lite only when it is weakly associated in a drugRBC mem-
section. brane protein complex (drugRBC membrane protein
1. Drug adsorption: The patient produces an IgG antibody to immunogenic complex mechanism). The antibodies acti-
a drug. When the drug is taken by the patient, the drug vate complement and trigger acute intravascular hemolysis
binds strongly to the patients RBCs. The IgG drug antibody that may progress to renal failure.2 Hemolysis occurs
binds to the drug attached to the RBCs, usually without abruptly after short periods of drug exposure or upon read-
complement activation. Because the offending antibody ministration of the drug.22 Examples of drugs that elicit
is IgG and is strongly attached to the RBCs via the drug, antibodies in this category are piperacillin, quinidine, and
hemolysis is extravascular by splenic macrophages, which ceftriaxone.22 Laboratory features include a positive DAT
remove the antibody- and drug-coated RBCs from the reaction with anti-C3d and occasionally with anti-IgG. In
circulation. the indirect AHG test, the patients serum reacts with
2. DrugRBC membrane protein immunogenic complex: A drug untreated, normal RBCs only in the presence of the drug.
binds loosely to an RBC membrane protein to form a drug Because C3d is the predominant protein on the RBCs, the
RBC protein immunogenic complex or epitope. The patient result of the indirect AHG test on an eluate of the patients
produces an IgM and/or IgG antibody that binds to the RBC is usually negative.2,6
complex on the RBCs and complement is fully activated,
which causes acute intravascular hemolysis. Drug-Independent Antibodies
3. RBC autoantibody induction: A drug induces the patient to Drug-independent antibodies are IgG, warm-reactive, RBC auto-
produce IgG warm-reactive autoantibodies against RBC self- antibodies induced by the drug (RBC autoantibody induction
antigens. These autoantibodies react at 37 C, and the labo- mechanism). These autoantibodies have the same serologic
ratory findings are indistinguishable from those in WAIHA. reactivity as those causing WAIHA, and they do not require the
Hemolysis is extravascular and is mediated by macrophages presence of the drug for in vitro reactivity. Hemolysis is extra-
predominantly in the spleen. vascular, mediated by macrophages predominantly in the
Several authors have suggested that all drug-induced spleen, usually with a gradual onset of anemia. Examples of
immune hemolysis is explained by a single mechanism, known drugs that elicit antibodies in this category are fludarabine,
as the unifying theory. This theory proposes that a drug interacts methyldopa, and procainamide.22,24 Laboratory features include
with the RBC membrane and generates multiple immunogenic a positive DAT reaction with anti-IgG. In the indirect AHG test,
epitopes that can elicit an immune response to (1) the drug the patients serum and an eluate of the patients cells generally
alone, (2) the drugRBC membrane protein combination, or react at 37 C with all screening and panel RBCs and with the
(3) an RBC membrane protein alone.24,26 patients own RBCs.2,6

Antibody Characteristics Nonimmune Drug-Induced Hemolysis


Antibodies implicated in DIIHA can be divided into two In drug-induced nonimmunologic protein adsorption, the patient
general types: drug-dependent (most common) and drug- does not produce an antibody to the drug or to RBCs. The
independent antibodies.2,24 Some drugs are able to induce a mechanism is also called the membrane modification method,
combination of both type of antibodies.22,27,28 because certain drugs such as high-dose cephalosporins and
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 361

cisplatin can alter the RBC membrane so that numerous pro- The immediate investigation of a suspected HTR includes
teins, including IgG and complement, adsorb on the RBC (1) a clerical check for errors, (2) examination of a posttransfu-
surface.24,27 This phenomenon results in a positive DAT finding, sion specimen for hemolysis, and (3) performance of the DAT
but only rarely has hemolysis been reported. The indirect AHG on the posttransfusion specimen.29 If an AHTR occurred,
test on the patients serum and an eluate of the patients RBCs hemoglobinemia and hemoglobinuria are detectable, and the
yields negative results.2 DAT result is positive. DAT findings may be negative, however,
if all the donor cells are lysed.29 The hemoglobin and serum
Treatment haptoglobin levels decrease, but the serum indirect bilirubin
After a DIIHA is recognized and confirmed, the first treatment will not begin to rise until 2 to 3 days after the episode. The
is to discontinue the drug. Most patients will gradually show ABO and Rh typing, antibody screen, and cross-matching are
improvement within a few days to several weeks.6,22 In cases in repeated on the recipient and the donor blood to identify the
which a warm-reacting autoimmune antibody is present, the blood group incompatibility. Coagulation tests reveal and
positive DAT result may persist for months after a hematologic assess the risk of DIC.
recovery.22 If the anemia is severe, the patient may require
RBC transfusion or plasma exchange.22 Regardless of mecha- Delayed Hemolytic Transfusion Reaction
nism, future episodes of DIIHA are prevented by avoidance of A delayed hemolytic transfusion reaction (DHTR) may occur
the drug. days to weeks after transfusion as the titer of alloantibodies
increases.31 Often, the patient has been alloimmunized by a
pregnancy or previous transfusion, but the antibody titer was
ALLOIMMUNE HEMOLYTIC ANEMIAS
below the level of serologic detection at the time of transfusion.
Hemolytic Transfusion Reaction The second exposure to the antigen results in an increase in
One of the most severe and potentially life-threatening com- titer (anamnestic response). The antibody is usually IgG, is
plications of blood transfusion is a hemolytic transfusion reac- reactive at 37 C, and may or may not be able to partially or
tion (HTR) due to immune-mediated destruction of donor fully activate complement. The antibodies most often impli-
cells by an antibody in the recipient. The offending antibody cated in DHTRs are directed against the following antigens: Jka,
in the recipient may be IgM or IgG, complement may be par- Jkb, E, C, c, K, Fya, S, and s.31 The patients antibody binds to
tially or fully activated or not activated at all, and the hemolysis the transfused RBCs, which leads to extravascular hemolysis,
may be intravascular or extravascular depending on the with or without complement activation. Inadequate posttrans-
characteristics of the antibody. HTRs can have an acute or fusion hemoglobin response, positive DAT results for IgG
delayed onset. and/or C3d, morphologic evidence of hemolysis, and indirect
bilirubinemia are the principal signs.31
Acute Hemolytic Transfusion Reaction
Acute hemolytic transfusion reactions (AHTRs) occur within Hemolytic Disease of the Fetus and Newborn
minutes to hours of the initiation of a transfusion.29 The most Hemolytic disease of the fetus and newborn (HDFN) occurs
common cause of AHTR is the accidental transfusion of ABO- when an IgG alloantibody produced by the mother crosses the
incompatible donor red cells into a recipient. An example is the placenta into the fetal circulation and binds to fetal RBCs that
transfusion of group A red cells into a group O recipient. The are positive for the corresponding antigen. The IgG-sensitized
recipient has preformed, naturally occurring anti-A (IgM) that fetal RBCs are cleared from the circulation by macrophages in
is capable of fully activating complement to C9 upon binding the fetal spleen, and an anemia gradually develops. There is
to the A antigen on donor red cells. There is rapid, complement- erythroid hyperplasia in the fetal bone marrow and extramed-
mediated intravascular hemolysis and activation of the coagu- ullary erythropoiesis in the fetal spleen, liver, kidneys, and
lation system. ABO-incompatible transfusions are usually due adrenal glands.32 Many nucleated RBCs are released into the
to clerical error and have been estimated to occur in approxi- fetal circulation. If the anemia is severe in utero, it can lead to
mately 1 in 38,000 to 1 in 70,000 RBC transfusions.29 The generalized edema, ascites, and a condition called hydrops
severity of AHTR is variable and is affected by the infusion rate fetalis, which is fatal if untreated.32,33
and volume of blood transfused.30 AHTR carries an estimated In Rh HDFN an Rh(D)-negative mother has preformed
mortality rate of 2%.29 AHTRs can occur due to incompatibili- anti-D antibodies (IgG, reactive at 37 C) from exposure to the
ties involving other blood group systems, but these are rare.29 D antigen either through immunization in a previous preg-
Symptoms of severe intravascular hemolysis found in ABO- nancy with a D-positive baby or from previous transfusion of
related AHTRs begin within minutes or hours and may include blood products with D-positive RBCs. In subsequent pregnan-
chills, fever, urticaria, tachycardia, nausea and vomiting, chest cies, the anti-D (IgG) crosses the placenta, and if the fetus is
and back pain, shock, anaphylaxis, pulmonary edema, conges- D positive, the anti-D binds to D antigen sites on the fetal
tive heart failure, and bleeding due to disseminated intra RBCs. These anti-Dsensitized fetal RBCs are cleared from the
vascular coagulation (DIC). The transfusion is immediately circulation by extravascular hemolysis in the fetal spleen, and
terminated. Treatment is urgent and includes an effort to anemia and hyperbilirubinemia develop. During pregnancy,
prevent or correct shock, maintain renal circulation, and the indirect bilirubin produced by the fetus passes across the
control the DIC. placenta and is excreted by the mother. After delivery, however,
362 PART IV Hematopathology: Erythrocyte Disorders

the infants immature liver cannot conjugate the indirect bili- TABLE 25-3 Characteristics of Rh and ABO Hemolytic
rubin efficiently, and as the antibody-mediated destruction of Disease of the Fetus and Newborn
newborn RBCs continues, the serum indirect bilirubin accumu-
Rh ABO
lates. If excessive indirect bilirubin passes the blood-brain
barrier, permanent brain damage due to bilirubin encepha- Blood groups
lopathy (also called kernicterus) can occur. Consequently, the Mother Rh (D) negative O
hyperbilirubinemia, along with the anemia, is closely moni- Child Rh (D) positive A or B
tored in the neonate. Severity of disease Severe Mild
Jaundice Severe Mild
Laboratory Findings Spherocytes on Rare Usually present
During the first trimester of pregnancy, ABO and Rh typing is peripheral blood film
performed on the mothers blood along with an indirect AHG Anemia Severe If present, mild
test for antibodies in her serum.33 An unimmunized D-negative Direct antiglobulin test Positive Negative or weakly positive
mother receives Rh immune globulin at 28 weeks gestation result
and again within 72 hours of delivery of a D-positive infant to
prevent alloimmunization to the D antigen.33 Rh-negative
women who experience spontaneous or induced abortion also
receive Rh immune globulin. If the mother is alloimmunized prevent hydrops fetalis.32,33 After delivery, the neonate may
to the D antigen, the titer of anti-D is determined by the indi- need exchange transfusions and phototherapy to reduce the
rect AHG test. Antibody titers are repeated at 18 weeks gesta- level of serum indirect bilirubin and prevent kernicterus.
tion and every 2 to 4 weeks thereafter until delivery. Titration
of the antibody does not predict the severity of HDFN; rather, Hemolytic Disease of the Fetus and Newborn
it helps determine when to monitor for HDFN by additional Caused by Other Blood Group Antigens
methods, such as spectrophotometric analysis of amniotic ABO HDFN is more common than Rh disease and may occur
fluid bilirubin.34 Amniocentesis is accurate at predicting severe during the first pregnancy. Unlike Rh disease, ABO disease is
fetal anemia, but it is an invasive procedure and carries some asymptomatic or produces mild hyperbilirubinemia and
risk of fetal loss. If severe fetal anemia and HDFN due to anti-D anemia. ABO HDFN is seen in some infants with group A
is suspected, a percutaneous umbilical fetal blood sample can or B blood born to group O mothers who, in addition to the
be obtained and tested for the hemoglobin level to determine usual IgM ABO antibodies, produce IgG anti-A and anti-B,
the severity of the anemia.32 which are capable of crossing the placenta. The disease is
At delivery, testing for the newborn is performed on milder than Rh HDFN, because most of the maternal anti-A
umbilical cord blood.33 Neonates with Rh HDFN have a and anti-B antibodies are the IgM isotype and do not cross the
decreased hemoglobin level, increased reticulocyte count, and placenta.32
increased level of serum indirect bilirubin. The peripheral The DAT result for the newborn with ABO HDFN is only
blood film shows polychromasia and many nucleated RBCs. weakly positive and may be negative. Spherocytes and poly-
ABO (only forward grouping) and Rh typing as well as the chromasia on the peripheral blood film are typical.32 Table
DAT are also performed. The DAT result shows positivity 25-3 presents a comparison of HDFN caused by ABO and Rh
for IgG, and anti-D can be demonstrated in an eluate of the incompatibility.
infants RBCs.33 HDFN can be caused by other IgG antibodies, particularly
antibodies to the K, c, and Fya antigens.33 HDFN due to other
Treatment for the Affected Infant blood group antibodies is rare.33 Varying degrees of anemia,
Treatment for a fetus affected by HDFN may include intrauter- jaundice, and kernicterus are the adverse clinical outcomes in
ine transfusion, which can be used to correct fetal anemia and all forms of HDFN.

SUMMARY
The immune hemolytic anemias are classified into autoimmune Hemolysis mediated by IgG occurs with or without complement
hemolytic anemia, drug-induced immune hemolytic anemia, and activation; IgG-sensitized RBCs are removed from the circulation
alloimmune hemolytic anemia. by macrophages in the spleen; partial phagocytosis produces
Hemolysis mediated by IgM requires complement; hemolysis may spherocytes, which are trapped in the spleen and phagocytized;
be extravascular (mainly in the liver) if complement is partially acti- IgG- and C3b-sensitized RBCs are removed by macrophages in
vated to C3b, or intravascular if complement is fully activated to C9. the spleen and liver.
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 363

Laboratory findings in immune hemolytic anemia include (3) an RBC membrane protein only. In vitro reactions of antibodies
decreased hemoglobin level, increased reticulocyte count, in DIIHA may be drug dependent or drug independent.
increased levels of serum indirect bilirubin and lactate dehydro- AHTRs occur within minutes to hours after the start of an RBC
genase, and decreased serum haptoglobin level. The peripheral transfusion and most often involve transfusion of ABO-incompatible
blood film may show polychromasia, spherocytes (IgG-mediated blood; the hemolysis is predominantly intravascular. DHTRs
hemolysis), or RBC agglutination (cold agglutinins). The DAT may occur days or weeks after the transfusion and represent an
detects in vivo sensitization of RBCs by IgG and/or C3d. anamnestic response to a donor antigen; the hemolysis is
The classification of autoimmune hemolytic anemia includes extravascular.
WAIHA, CAD, PCH, and mixed-type autoimmune hemolytic anemia. HDFN occurs when an IgG alloantibody produced by the mother
WAIHA is the most common form of autoimmune hemolytic anemia crosses the placenta into the fetal circulation and binds to fetal
and involves IgG autoantibodies with optimum reactivity at 37 C. RBCs that are positive for the corresponding antigen. The IgG-
The anemia varies from mild to severe, and characteristic mor- sensitized fetal RBCs are cleared from the circulation by macro-
phologic features on the peripheral blood film are polychromasia phages in the fetal spleen, and an anemia gradually develops; the
and spherocytes. usual laboratory findings in the neonate are anemia, hyperbiliru-
CAD is caused by an IgM autoantibody with optimum reactivity at binemia, and a positive DAT result.
4 C and a thermal amplitude of greater than 30 C. RBC agglu- ABO HDFN is more common than Rh HDFN and produces no
tination may be observed on a peripheral blood film, and aggluti- symptoms or mild anemia. Rh HDFN due to anti-D results in severe
nates may cause interference with the CBC analysis on automated anemia; administration of Rh immune globulin at 28 weeks gesta-
instruments. tion and after delivery of a D-positive infant prevents immunization
PCH is due to a biphasic IgG autoantibody with anti-P specificity. to the D antigen in D-negative pregnant women.
The antibody binds to the P antigen on the RBCs and partially
activates complement at 4 C; complete complement activation Now that you have completed this chapter, go back and
and hemolysis occurs upon warming the sample to 37 C. read again the case study at the beginning and respond
In DIIHA, the patient produces antibodies to (1) a drug only, (2) a to the questions presented.
complex of a drug loosely bound to an RBC membrane protein, or

R E V I E W Q UESTIONS
1. The pathophysiology of immune hemolysis with IgM anti- 4. The antibody that mediates the hemolytic process in
bodies always involves: WAIHA is usually of which immunoglobulin class?
a. Complement a. A
b. Autoantibodies b. E
c. Abnormal hemoglobin molecules c. G
d. Alloantibodies d. M

2. In hemolysis mediated by IgG antibodies, which abnor- 5. The hemolysis in PCH is usually:
mal RBC morphology is typically observed on the periph- a. Extravascular with complement activation to C3b
eral blood film? b. Extravascular with no complement activation
a. Spherocytes c. Intravascular with complement activation to C9
b. Schistocytes d. Not significant
c. Rouleaux
d. RBC agglutination 6. The difference between nonpathologic and pathologic
cold agglutinins is that the pathologic types:
3. The most important finding in the diagnostic investigation a. Are IgG and react optimally at 37 C
of a suspected autoimmune hemolytic anemia is: b. Have a thermal amplitude greater than 30 C
a. Detection of a low hemoglobin and hematocrit c. Readily activate complement
b. Observation of hemoglobinemia in a specimen d. Are only found in the elderly
c. Recognition of a low reticulocyte count
d. Demonstration of IgG, C3d, or both on the RBC 7. Which one of the following statements is true about DHTR:
surface a. It is usually due to an ABO incompatibility
b. Hemoglobinemia and hemoglobinuria frequently occur
c. It is due to an anamnestic response after repeat
exposure to a blood group antigen
d. The DAT yields a positive result for C3d only
364 PART IV Hematopathology: Erythrocyte Disorders

8. Chronic secondary CAD is most often associated with: 10. A Group A Rh-negative mother gave birth to a Group O
a. Antibiotic therapy Rh-positive baby. The baby is at risk for HDFN if:
b. M. pneumoniae infection a. This was the mothers first pregnancy
c. B-cell malignancies b. The mother has IgG ABO antibodies
d. Infectious mononucleosis c. The mother was previously immunized to the D
antigen
9. A 63-year-old man is being evaluated because of a decrease d. The mother received Rh immune globulin prior to
in hemoglobin of 5 gm/dL after a second cycle of fludara- delivery
bine for treatment of chronic lymphocytic leukemia. The
patients DAT result is strongly positive for IgG only, and
antibody testing on his serum and an eluate of his RBCs
yields positive results with all panel cells and the patients
own cells. This suggests which mechanism of immune
hemolysis for this patient?
a. Drug-RBC membrane protein complex
b. Drug adsorption
c. RBC autoantibody induction
d. Drug-induced nonimmunologic protein adsorption

REFERENCES
1. Powers A, Silberstein LE: Autoimmune hemolytic anemia. In 14. Berentsen S, Ulvestad E, Gjertsen BT, et al: Rituximab for
Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology: primary chronic cold agglutinin disease: a prospective study
basic principles and practice, ed 5, Philadelphia, 2009, Churchill of 37 courses of therapy in 27 patients. Blood 103:2925-2928,
Livingstone. 2004.
2. Leger RM: The positive direct antiglobulin test and immune- 15. Schollkopf C, Kjeldsen L, Bjerrum OW, et al: Rituximab in
mediated hemolysis. In Roback JD, Combs MR, Grossman BJ, chronic cold agglutinin disease: a prospective study of 20
et al, editors: Technical manual, ed 16, Bethesda, Md, 2008, patients. Leuk Lymphoma 47:253-260, 2006.
American Association of Blood Banks. 16. Berentsen S, Randen U, Vagan AM, et al: High response rate
3. Mandle R, Barrington RA, Carroll MC: Complement and and durable remissions following fludarabine and rituximab
immunoglobulin biology. In Hoffman R, Benz EJ, Jr, Shattil combination therapy for chronic cold agglutinin disease.
SJ, et al, editors: Hematology: basic principles and practice, ed 5, Blood Epub July 15, 2010.
Philadelphia, 2009, Churchill Livingstone. 17. Sokol RJ, Hewitt S, Stamps BK, et al: Autoimmune haemolysis
4. Prodinger WM, Wrzner R, Stoiber H, et al: Complement. In in childhood and adolescence. Acta Haematol 72:245-257,
Paul WE, editor: Fundamental immunology, ed 5, Philadelphia, 1984.
2003, Lippincott Williams & Wilkins. 18. Gottsche B, Salama A, Mueller-Eckhardt C: Donath-
5. Friedberg RC, Johari VP: Autoimmune hemolytic anemia. In Landsteiner autoimmune hemolytic anemia in children. A
Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes study of 22 cases. Vox Sang 58:281-286, 1990.
clinical hematology, ed 12, Philadelphia, 2009, Lippincott 19. Gertz MA. Management of cold haemolytic syndrome. Br J
Williams & Wilkins. Haematol 138:422-429, 2007.
6. Gehrs BC, Friedberg RC: Autoimmune hemolytic anemia. Am 20. Mayer B, Yurek S, Kiesewetter H, et al: Mixed-type
J Hematol 69:258-271, 2002. autoimmune hemolytic anemia: differential diagnosis and
7. Bellia M, Georgopoulos J, Tsevrenis V, et al: The investigation critical review of reported cases. Transfusion 48:2229-2234,
of the significance of a positive direct antiglobulin test in 2008.
blood donors. Immunohematology 18:78-81, 2002. 21. Sokol RJ, Hewitt S, Stamps BK: Autoimmune haemolysis: an
8. Stratton F, Rawlinson VI, Merry A, et al: Positive direct anti- 18 year study of 865 cases referred to a regional transfusion
globulin test in normal individuals. Clin Lab Haematol 5:17- centre. Br Med J 282:2023-2027, 1981.
21, 1983. 22. Garratty G: Immune hemolytic anemia associated with drug
9. Garratty G, Arndt P, Domen R, et al: Severe autoimmune therapy. Blood Rev 24:143-150, 2010.
hemolytic anemia associated with IgM warm autoantibodies 23. Johnson ST, Fueger JT, Gottschall JL: One centers experience:
directed against determinants on or associated with gly- the serology and drugs associated with drug-induced hemo-
cophorin A. Vox Sang 72:124-130, 1997. lytic anemiaa new paradigm. Transfusion 47:697-702,
10. Dhingra KK, Jain D, Mandal S, et al: Evans syndrome: a study 2007.
of six cases with review of literature. Hematology 13:356-360, 24. Garratty G: Drug-induced immune hemolytic anemia. Hema-
2008. tology Am Soc Hematol Educ Program, pp 73-79, 2009.
11. Berentsen S, Beiske K, Tjonnfjord GE: Primary chronic cold 25. Garratty G, Arndt PA: An update on drug-induced
agglutinin disease: an update on pathogenesis, clinical fea- immune hemolytic anemia. Immunohematology 23:105-119,
tures and therapy. Hematology 12:361-370, 2007. 2007.
12. Petz LD: Cold antibody autoimmune hemolytic anemias. 26. Habibi B: Drug induced red blood cell autoantibodies
Blood Rev 22:1-15, 2008. co-developed with drug specific antibodies causing haemo-
13. Berentsen S, Ulvestad E, Langholm R, et al: Primary chronic lytic anemias. Br J Haematol 61:139-143, 1985.
cold agglutinin disease: a population based clinical study of 27. Arndt P, Garratty G, Isaak E, et al: Positive direct and indirect
86 patients. Hematologica 91:460-466, 2006. antiglobulin tests associated with oxaliplatin can be due
CHAPTER 25 Extrinsic Defects Leading to Increased Erythrocyte DestructionImmune Causes 365
to drug antibody and/or drug-induced nonimmunologic 32. Ramasethu J, Luban NL: Alloimmune hemolytic disease of
protein adsorption. Transfusion 49:711-718, 2009. the fetus and newborn. In Kaushansky K, Lichtman MA,
28. Salama A: Drug-induced immune hemolytic anemia. Expert Beutler TJ, et al, editors: Williams hematology, ed 8, New York,
Opin Drug Saf 8:73-79, 2009. 2010, McGraw-Hill Medical, chap 54. Available at: http://
29. Lerner NB, Refaai MA, Blumberg N: Red cell transfusion. www.accessmedicine.com/content.aspx?aID=6131386.
In Kaushansky K, Lichtman MA, Beutler TJ, et al, editors: Accessed August 9, 2010.
Williams hematology, ed 8, New York, 2010, McGraw-Hill 33. Kennedy MS: Perinatal issues in transfusion practice. In
Medical, chap 140. Available at: http://accessmedicine.com/ Roback JD, Combs MR, Grossman BJ, et al, editors: Technical
content.aspx?aID=6236286. Accessed August 14, 2010. manual, ed 16, Bethesda, Md, 2008, American Association of
30. Eder AF, Chambers LA: Noninfectious complications Blood Banks.
of blood transfusion. Arch Pathol Lab Med 131:708-718, 34. Sikkel E, Vandenbussche FPHA, Oepkes D, et al: Amniotic
2007. fluid delta OD 450 values accurately predict severe fetal
31. Perrotta PL, Snyder EL: Noninfectious complications of trans- anemia in D-alloimmunization. Obstet Gynecol 100:51-57,
fusion therapy. Blood Rev 15:69-83, 2001. 2002.
26 (Structural
Hemoglobinopathies
Defects in Hemoglobin)
Tim R. Randolph

OUTLINE OBJECTIVES
Structure of Globin Genes After completion of this chapter, the reader will be able to:
Hemoglobin Development
Genetic Mutations 1. Compare and contrast structural hemoglobin (Hb) 12. Define the treatment goal for Hb S disease and
Zygosity disorders with thalassemias. discuss various treatments and their purposes.
Pathophysiology 2. Briefly describe globin gene structure and the devel- 13. Identify the amino acid substitution and electropho-
Nomenclature opment of normal human hemoglobins throughout retic mobility of Hb C and Hb C-Harlem.
Hemoglobin S prenatal and postnatal life. 14. Describe the amino acid substitution in Hb E and the
Hemoglobin C 3. Characterize the categories of the molecular abnor- importance of genetic counseling.
Hemoglobin C-Harlem malities found in the various hemoglobinopathies. 15. Explain the importance of and methods for differen-
(Hemoglobin 4. Differentiate between homozygous and heterozygous tiating Hb D and Hb G from Hb S.
C-Georgetown) states. 16. Describe the laboratory findings in patients who are
Hemoglobin E
5. Define the pathologic basis of sickle cell disorder. compound heterozygotes for Hb S and Hb D, Hb
Hemoglobin O-Arab
6. Describe the inheritance pattern of Hb S. O-Arab, Hb Korle Bu, or Hb C-Harlem.
Hemoglobin D and
Hemoglobin G 7. State the amino acid substitution found in Hb S and 17. Identify the causes of methemoglobinemia.
Compound Heterozygosity describe the electrophoretic mobility of Hb S. 18. Describe the inheritance patterns and causes of
with Hemoglobin S and 8. Describe the solubility of Hb S in the deoxygenated unstable hemoglobins.
Another -Hemoglobin state. 19. Discuss hemoglobins with increased and decreased
Mutation 9. Discuss the clinical presentation of sickle cell disease, oxygen affinities, and explain how they differ from
Hemoglobin M including differentiation among types of sickle cell crises. unstable hemoglobins.
Unstable Hemoglobin 10. Locate the geographic region, define the frequency 20. Predict possible inheritance of hemoglobinopathies
Variants of occurrence, and describe the impact of the from the genotype of parents and the inheritance
Hemoglobins with incidence of Plasmodium falciparum malaria and pattern for the gene.
Increased and
glucose-6-phosphate dehydrogenase deficiency on 21. Interpret patient hemoglobin electrophoresis patterns on
Decreased Oxygen
hemoglobin variants. cellulose acetate or agarose gel at pH 8.4 and on citrate
Affinity
11. Describe the peripheral blood cell analysis, chemis- agar at pH 6.0 to 6.2 when given control patterns.
try panels, and other laboratory procedures used in 22. Correlate hemoglobin electrophoresis and solubility
the diagnosis of hemoglobinopathies. test results to identify hemoglobinopathies.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

An 18-year-old African American woman was seen in the Patient Results Reference Range
emergency department for fever and abdominal pain. The 9
Platelets ( 10 /L) 410 150-450
following results were obtained on a complete blood count: Segmented neutrophils (%) 75 50-70
Lymphocytes (%) 18 18-42
Patient Results Reference Range Monocytes (%) 3 2-11
9
WBCs ( 10 /L) 11.9 4.5-11.0 Eosinophils (%) 3 1-3
RBCs ( 1012/L 3.67 4.00-5.40 Basophils (%) 1 0-2
Hb (g/dL) 10.9 12.0-15.0 Reticulocytes (%) 3.1 0.5-1.5
Hct (%) 32.5 35-49 Continued
RDW (%) 19.5 11.5-14.5

366
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 367

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

A typical field in the patients peripheral blood film is shown


in Figure 26-1.
Electrophoresis on cellulose acetate at alkaline pH showed
50.9% Hb S and 49.1% Hb C.
1. Select confirmatory tests that should be performed and
describe the expected results.
2. Describe the characteristic RBC morphology on the
peripheral blood film.
3. Based on the electrophoresis and RBC morphology results,
what diagnosis is suggested?
4. If this patient were to marry a person of genotype Hb AS,
what would be the expected frequency of genotypes for
each of four children?

Figure 26-1 Peripheral blood film for the patient in the case study
(1000). (Courtesy Ann Bell, University of Tennessee, Memphis.)

H
emoglobinopathy refers to a disease state (opathy) the globin genes, and , are located on chromosome 16 and
involving the hemoglobin (Hb) molecule. All hemo- are referred to as -like genes. The remaining four globin genes,
globinopathies result from a genetic mutation in one , , , and , are located on chromosome 11 and are referred
or more genes that affect hemoglobin synthesis. The genes that to as -like genes. In the human genome, there is one copy of
are mutated can code for either the proteins that make up the each globin gene per chromatid for a total of two genes per
hemoglobin molecule (globin chains) or the proteins involved person with the exception of and . There are two copies of
in synthesizing or regulating synthesis of the globin chains. the and genes per chromatid for a total of four genes per
Regardless of the mutation encountered, all hemoglobinopa- person. Each globin gene codes for the corresponding globin
thies affect hemoglobin synthesis in one of two ways: qualita- chain: the -globin gene is used as the template to synthesize
tively or quantitatively. In qualitative hemoglobinopathies, the -globin chain, the -globin gene codes for the -globin
hemoglobin synthesis occurs at a normal or near-normal rate, chain, and so forth.
but the hemoglobin molecule has an altered amino acid
sequence within the globin chains. This change in amino acid
sequence alters the structure of the hemoglobin molecule
HEMOGLOBIN DEVELOPMENT
(structural defect) and its function (qualitative defect). In
contrast, thalassemias result in a reduced rate of hemoglobin Each human hemoglobin molecule is composed of four globin
synthesis (quantitative) but do not affect the amino acid chains, a pair of -like chains and a pair of -like chains.
sequence of the globin chains. A reduction in the amount of During the first 3 months of embryonic life, only one -like
hemoglobin synthesized produces an anemia and stimulates gene () and one -like gene () are activated, which results
the production of other hemoglobins not affected by the muta- in the production of and chains that pair to form a hemo-
tion in an attempt to compensate for the anemia. Based on this globin type called Gower-1 (22). Shortly thereafter, and
distinction, hematologists divide hemoglobinopathies into chain synthesis begins, which leads to the production of Hb
two categories: structural defects (qualitative) and thalassemias Gower-2 (22) and Hb Portland (22). Later in fetal develop-
(quantitative). To add confusion to the classification scheme, ment, and synthesis ceases; this leaves and chains, which
many hematologists also refer to only the structural defects as pair to produce Hb F (22), also known as fetal hemoglobin.
hemoglobinopathies. This chapter describes the structural or During the 6 months after birth, chain synthesis gradually
qualitative defects that are referred to as hemoglobinopathies; the decreases and is replaced by chain synthesis, so that Hb
quantitative defects (thalassemias) are described in Chapter 27. A (22), also known as adult hemoglobin, is produced. The
remaining globin gene, , becomes activated around birth,
producing chains at low levels that pair with chains to
STRUCTURE OF GLOBIN GENES
produce the second adult hemoglobin, called Hb A2 (22).
As discussed in Chapter 10, there are six functional human Normal adults produce Hb A (95%), Hb A2 (less than 3.5%),
globin genes located on two different chromosomes. Two of and Hb F (less than 1% to 2%).
368 PART IV Hematopathology: Erythrocyte Disorders

TABLE 26-1 Molecular Abnormalities of Hemoglobin Chain extensions occur when the stop codon is mutated,
Variants so that translation continues beyond the typical last codon.
Amino acids continue to be added until a stop codon is reached
NUMBER OF VARIANTS
by chance. This process produces globin chains that are longer
Total
than normal. Significant globin chain extensions usually result
Amino acid substitution 373 588 67 100 1128 in degradation of the globin chain and a quantitative defect. If
Deletions or insertions 45 159 5 8 217 the extension of the globin chain is insufficient to produce
Fusions 0 8 7 1 16 significant degradation, however, the defect is qualitative and
Total* 418 755 79 109 1361 is classified as a hemoglobinopathy. Hemoglobin molecules
with extended globin chains fold inappropriately, which affects
Data from Patrinos GP, Giardine B, Riemer C, etal: Improvements in the HbVar data- hemoglobin structure and function.
base of human hemoglobin variants and thalassemia mutations for population and
sequence variation studies, Nucl Acids Res 32(database issue):D537-541, 2004. Gene fusions occur when two normal genes break between
Available at: http://globin.cse.psu.edu/hbvar/menu.html. Accessed May 3, 2010. nucleotides, switch positions, and anneal to the opposite gene.
*The table is designed to provide a relative distribution of mutation types and includes For example, if a gene and a gene break in similar locations,
thalassemias and structural variants. Mutations are being added regularly, and at the
time of publication there were 1034 hemoglobin variants and 395 thalassemias. The switch positions, and reanneal, the resultant genes would be
total number of entries in the table (1361) is less than the sum of the hemoglobin and fusion genes in which the head of the fusion gene
variants and thalassemias because some mutants are listed in both categories. is from one original gene and the tail is from the other. As
long as the reading frames are not disrupted and the globin
chain lengths are similar, the genes are transcribed and trans-
GENETIC MUTATIONS
lated into hybrid globin chains. The fusion chains fold differ-
More than 1000 structural hemoglobin variants (hemoglobin- ently, however, and affect the corresponding hemoglobin
opathies) are known to exist throughout the world, and more function. Sixteen fusion globin chains have been identified
are being discovered regularly (Table 26-1).1 Each of these (see Table 26-1).
hemoglobin variants results from one or more genetic muta-
tions that alter the amino acid sequence. Some of these changes
ZYGOSITY
alter the molecular structure of the hemoglobin molecule,
ultimately affecting hemoglobin function. The types of genetic Zygosity refers to the association between the number of gene
mutations that occur in the hemoglobinopathies include mutations and the level of severity of the resultant genetic
point mutations, deletions, insertions, chain extensions, and defect. Generally, there is a level of severity associated with each
fusions involving one or more of the adult globin genes, gene that is normally used to synthesize the protein product.
, , and .2 For the normal adult globin genes, there are four copies of the
Point mutation is the most common type of genetic muta- and genes and two copies of the and genes. In theory,
tion occurring in the hemoglobinopathies. Point mutation is this could result in four levels of severity for and mutations
the replacement of one original nucleotide in the normal gene and two levels of severity for the and mutations. Expressed
with a different nucleotide. Because one nucleotide is replaced another way, if all things were equal, it would require twice as
by one nucleotide, the codon triplet remains intact, and the many mutations within the and genes to produce the same
reading frame is unaltered. This results in the substitution of physiologic effect as mutations within the and genes.
one amino acid in the globin chain product at the position Because the and genes are transcribed and translated at such
corresponding to the location of the original point mutation. low levels in adults, however, mutations of either gene would
As can be seen in Table 26-1, 1128 of the 1361 known hemo- have little impact on overall hemoglobin function. In addition,
globin variants result from a point mutation that causes an because the dominant hemoglobin in adults, Hb A, is com-
amino acid substitution. It also is possible to have two point posed of and chains, gene mutations would affect overall
mutations occurring in the same globin gene, which results in hemoglobin function to a greater extent than the same number
two amino acid substitutions within the same globin chain. of gene mutations. This partially explains the greater number
Over 30 mutations occur by this mechanism.2 of identified chain variants compared with chain variants,
Deletions involve the removal of one or more nucleotides, because a single mutation would be more likely to create a
whereas insertions result in the addition of one or more nucle- clinical condition than would a single mutation.
otides. Usually deletions and insertions are not divisible by The inheritance pattern of chain variants is referred to as
three and disrupt the reading frame, which leads to the nulli- heterozygous when one gene is mutated and homozygous when
fication of synthesis of the corresponding protein. This is the both genes are mutated. The terms disease and trait are also
case for the quantitative thalassemias (see Chapter 27). In commonly used to refer to the homozygous (disease) and
hemoglobinopathies, the reading frame usually remains intact, heterozygous (trait) states.
however; the result is the addition or deletion of one or more
amino acids in the globin chain product, which sometimes
PATHOPHYSIOLOGY
affects the structure and function of the hemoglobin molecule.
Of the 1361 variants described in Table 26-1, 217 variants Pathophysiology refers to the manner in which a disorder
result from deletions or insertions or both.2 translates into clinical symptoms. The impact of these point
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 369

mutations on hemoglobin function depends on the chemical most frequently occurring of the abnormal hemoglobins and
nature of the substituted amino acid, where it is located in the the most severe is Hb S.
globin chain, and the number of genes mutated (zygosity).
The charge and size of the substituted amino acid may alter
NOMENCLATURE
the manner in which the protein folds. A change in charge
affects the interaction of the substituted amino acid with adja- As hemoglobins were reported, they were designated by letters
cent amino acids. In addition, the size of the substituted amino of the alphabet. Normal adult hemoglobin and fetal hemoglo-
acid makes the protein either more or less bulky. Therefore, the bin were called Hb A and Hb F. By the time the middle of the
charge and the size of the substituted amino acid determine its alphabet was reached, however, it became apparent that
impact on hemoglobin structure by potentially altering the the alphabet would be exhausted before all mutations were
tertiary structure of the globin chain and the quaternary struc- named. Currently, some abnormal hemoglobins are assigned a
ture of the hemoglobin molecule. Changes in hemoglobin common designation and a scientific designation. The common
structure usually affect function. Location of the substitution name is selected by the discoverer and usually represents the
within the globin chain also has an impact on the degree of geographic area where the hemoglobin was identified. A single
structural alteration and hemoglobin function based on its capital letter is used to indicate a special characteristic of the
positioning within the molecule and the interactions with the hemoglobin variants, such as hemoglobins demonstrating
surrounding amino acids. In the case of the sickle cell muta- identical electrophoretic mobility but containing different
tion, the amino acid substitution results in hemoglobin poly amino acid substitutions, as in Hb G-Philadelphia, Hb
merization, leading to the formation of long hemoglobin G-Copenhagen, and Hb C-Harlem. The variant description also
crystals that stretch the RBC membrane and produce the char- can involve scientific designations that indicate the variant
acteristic crescent moon or sickle cell shape. chain, the sequential and the helical number of the abnormal
Zygosity also affects the pathophysiology of the disease. In amino acid, and the nature of the substitution. The designation
-hemoglobinopathies, zygosity predicts two severities of [6 (A3) GluVal] for the Hb S mutation indicates the substitu-
disease. In homozygous -hemoglobinopathies, in which both tion of valine for glutamic acid in the A helix in the chain at
genes are mutated, the variant hemoglobin becomes the the sixth position.1
dominant hemoglobin type, and normal hemoglobin (Hb A)
is absent. Examples are sickle cell disease (SCD, Hb SS) and
HEMOGLOBIN S
Hb C disease (Hb CC). In heterozygous -hemoglobinopathies,
one gene is mutated and the other is normal, which suggests Sickle Cell Anemia
a 50/50 distribution. In an attempt to minimize the impact of History
the abnormal hemoglobin, however, the variant hemoglobin Although the origin of sickle cell anemia has not been identi-
is usually present in lesser amounts than Hb A. Nevertheless, fied, symptoms of the disease have been traced in one Ghana-
in some cases they may be present in equal amounts. Examples ian family back to 1670.4 Sickle cell anemia was first reported
are Hb S trait (Hb AS) and Hb C trait (Hb AC). Patients with by a Chicago cardiologist, Herrick, in 1910 in a West Indian
homozygous SCD (Hb SS) inherit a severe form of the disease student with severe anemia. In 1917, Emmel recorded that
that occurs less frequently but requires lifelong medical inter- sickling occurred in nonanemic patients and in patients who
vention, which must begin early in life, whereas heterozygotes were severely anemic. In 1927, Hahn and Gillespie described
(Hb AS) are much more common but rarely experience the pathologic basis of the disorder and its relationship to the
symptoms. hemoglobin molecule. These investigators showed that sick-
Fishleder and Hoffman3 divided the structural hemoglobins ling occurred when a solution of red blood cells (RBCs) was
into four groups: (1) abnormal hemoglobins that result in deficient in oxygen and that the shape of the RBCs was revers-
hemolytic anemia, such as Hb S and the unstable hemoglobins; ible when that solution was oxygenated again.1,5 In 1946, Beet
(2) abnormal hemoglobins that result in methemoglobinemia, reported that malarial parasites were present less frequently in
such as Hb M; (3) hemoglobins with either increased or blood films from patients with SCD than in individuals without
decreased oxygen affinity; and (4) abnormal hemoglobins with SCD.6 It was determined that sickle cell trait confers a resistance
no clinical or functional effect. Imbalanced chain production against infection with Plasmodium falciparum occurring early in
also may be associated in rare instances with a structurally childhood between the time that passively acquired immunity
abnormal chain, such as Hb Lepore,1 because of the reduced dissipates and active immunity develops.7 In 1949, Pauling
production of the abnormal chain. The functional classification showed that when Hb S is subjected to electrophoresis, it
of selected hemoglobin variants is summarized in Box 26-1. migrates differently than does Hb A. This difference was shown
Many of the variants are clinically insignificant because they to be caused by an amino acid substitution in the globin chain.
do not show any physiologic effect. As discussed previously, Pauling and coworkers defined the genetics of the disorder and
most clinical abnormalities are associated with the chain clearly distinguished heterozygous sickle trait (Hb AS) from the
followed by the chain. Involvement of the and chains homozygous state (Hb SS).1
does occur, but because of the small amount of hemoglobin The term sickle cell diseases is used to describe a group of
involved, it is rarely detected and is usually of no consequence. symptomatic hemoglobinopathies that have in common sickle
Box 26-2 lists clinically significant abnormal hemoglobins. The cell formation and the associated crises. Patients with SCD are
370 PART IV Hematopathology: Erythrocyte Disorders

BOX 26-1 Functional Classification of Selected Hemoglobin (Hb) Variants

I. Homozygous: Hemoglobin Polymorphisms: c. Hb Providence: 2282Asn


The Variants That Are Most Common d. Hb Agenogi: 2290Lys
Hb S: 226Valsevere hemolytic anemia; sickling e. Hb Beth Israel: 22102Ser
Hb C: 226Lysmild hemolytic anemia f. Hb Yoshizuka: 22108Asp
Hb D-Punjab: 22121Glnno anemia
Hb E: 2226Lysmild microcytic anemia C. Unstable Hemoglobins
1. Hemoglobin may precipitate as Heinz bodies after splenectomy
II. Heterozygous: Hemoglobin Variants Causing (congenital Heinz body anemia)
Functional Aberrations or Hemolytic Anemia in a. Severe hemolysis: no improvement after splenectomy
the Heterozygous State Hb Bibba: 2136Pro2
A. Hemoglobins Associated with Methemoglobinemia Hb Hammersmith: 2242Ser
and Cyanosis Hb Bristol-Alesha: 2267Asp or 67Met
1. Hb M-Boston: 258Tyr2 Hb Olmsted: 22141Arg
2. Hb M-Iwate: 287Tyr2 b. Severe hemolysis: improvement after splenectomy
3. Hb M-Saskatoon: 2263Tyr Hb Torino: 243Val2
4. Hb M-Milwaukee-1: 2267Glu Hb Ann Arbor: 280Arg2
5. Hb M-Milwaukee-2 (Hyde Park): 2292Tyr Hb Genova: 2228Pro
6. Hb FM-Osaka: 2263Tyr Hb Shepherds Bush: 2274Asp
7. Hb FM-Fort Ripley: 2292Tyr Hb Kln: 2298Met
Hb Wien: 22130Asp
B. Hemoglobins Associated with Altered Oxygen c. Mild hemolysis: intermittent exacerbations
Affinity Hb Hasharon: 247His2
1. Increased affinity and polycythemia Hb Leiden: 226 or 7 (Glu deleted)
a. Hb Chesapeake: 292Leu2 Hb Freiburg: 2223 (Val deleted)
b. Hb J-Capetown: 292Gln2 Hb Seattle: 2270Asp
c. Hb Malmo: 2297Gln Hb Louisville: 2242Leu
d. Hb Yakima: 2299His Hb Zurich: 2263Arg
e. Hb Kempsey: 2299Asn Hb Gun Hill: 2291-95 (5 amino acids deleted)
f. Hb Ypsi (Ypsilanti): 2299Tyr d. No disease
g. Hb Hiroshima: 22146Asp Hb Etobicoke: 284Arg2
h. Hb Rainier: 22145Cys Hb Sogn: 2214Arg
i. Hb Bethesda: 22145His Hb Tacoma: 2230Ser
2. Decreased affinitymay have mild anemia or cyanosis 2. Tetramers of normal chains; appear in thalassemias
a. Hb Kansas: 22102Thr Hb Bart: 4
b. Hb Titusville: 294Asn2 Hb H: 4

From Henry JB: Clinical diagnosis and management by laboratory methods, ed 18, Philadelphia, 1991, Saunders, p 650. Originally modified from Winslow RM,
Anderson WF: The hemoglobinopathies. In Stanbury JB, Wyngaarden JB, Fredrickson DS, etal, editors: The metabolic basis of inherited disease, ed 5, New York,
1983, McGraw-Hill, chap 76. Updated from Patrinos GP, Giardine B, Riemer C, etal: Improvements in the HbVar database of human hemoglobin variants and thalas-
semia mutations for population and sequence variation studies, Nucl Acids Res 32(database issue):D537-541, 2004. Available at: http://globin.cse.psu.edu/hbvar/
menu.html. Accessed May 3, 2010.
Arg, Arginine; Asn, asparagine; Asp, aspartic acid; Cys, cysteine Gln, glutamine; Glu, glutamic acid; His, histidine; Leu, leucine; Lys, lysine; Met, methionine;
Pro, proline; Ser serine; Thr, threonine; Tyr, tyrosine; Val, valine.

either homozygous for Hb S (SS) or are compound heterozy- 16, whereas the -like genes (, , , and ) are located on the
gotes expressing Hb S in combination with another hemoglo- short arm of chromosome 11. With the exception of the
bin chain mutation like Hb C or -thalassemia. SCDs are the genes, which have four loci, each -like gene has two loci.
most common form of hemoglobinopathy, with Hb SS and -hemoglobin variants are inherited as autosomal codomi-
the variants Hb SC and Hb S-thalassemia (Hb S-thal) nants, with one gene inherited from each parent.1
occurring most frequently. Patients with SCD (Hb SS), Hb SC, or Hb S-thal have
inherited a sickle (S) gene from one parent and an S, C, or
Inheritance Pattern -thalassemia gene from the other. Among patients with SCD,
As stated earlier, the genes that code for the globin chains are individuals who are homozygotes (Hb SS) have more severe
located at specific loci on chromosomes 16 and 11. The -like disease than individuals who are compound heterozygotes for
genes ( and ) are located on the short arm of chromosome Hb S (Hb SC or Hb S-thal). Heterozygotes (Hb AS) are
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 371

A S S S A S
BOX 26-2 Clinically Important Hemoglobin (Hb)
Variants A AA AS A AS AS A AA AS
I. Sickle syndromes
A. Sickle cell trait (AS) A AA AS A AS AS S AS SS
B. Sickle cell disease
1. SS A B C
2. SC
3. SD-Los Angeles S S A S
4. SO-Arab
A AS AS A AA AS
5. S-Thalassemia
6. Shereditary persistence of fetal hemoglobin
S SS SS C AC SC
7. SE
II. Unstable hemoglobinscongenital Heinz body anemia (>200
variants) D E
III. Hemoglobins with abnormal oxygen affinity Figure 26-2 Punnett square illustrating the standard method for predict-
A. High affinityfamilial erythrocytosis (>115 variants) ing the inheritance of abnormal hemoglobins. Each parent contributes
B. Low affinityfamilial cyanosis (Hbs Kansas, Beth Israel, one gene.
Yoshizuka, Agenogi, Titusville, Providence)
IV. M hemoglobinsfamilial cyanosis (7 variants): Hb M-Boston, Hb Etiology and Pathophysiology
M-Iwate, Hb M-Saskatoon, Hb M-Milwaukee-1, Hb M-Milwaukee-2 Hb S is defined by the structural formula 226GluVal, which
(Hyde Park), Hb FM-Osaka, Hb FM-Fort Ripley indicates that on the chain at the sixth position, glutamic
V. Structural variants that result in a thalassemic phenotype acid is replaced by valine. Glutamic acid has a net charge of
A. -Thalassemia phenotype (1), whereas valine has a net change of (+0). This amino acid
1. Lepore Hbs ( fusion) substitution produces a change in charge of (+1), which affects
2. Hb E the electrophoretic mobility of the hemoglobin molecule. This
3. Hb-Indianapolis, Hb-Showa-Yakushiji, Hb-Geneva amino acid substitution also affects the way the hemoglobin
B. -Thalassemia phenotype chain termination mutants (e.g., Hb molecules interact with one another within the erythrocyte
Constant Spring) cytosol. The nonpolar (hydrophobic) valine amino acid has
Modified from Lukens JN: Abnormal hemoglobins: general principles (chap 39); been placed in the position that the polar glutamic acid once
Wong WC: Sickle cell anemia and other sickling syndromes (chap 40); Lukens held. Because glutamic acid is polar, the chain folds in such
JN: Unstable hemoglobin disease (chap 41). In Greer JP, Foerster J, Lukens JN, a way that glutamic acid extends outward from the surface of
etal, editors: Wintrobes clinical hematology, ed 11, Philadelphia, 2004, the hemoglobin tetramer to bind water and contribute to
Lippincott Williams & Wilkins.
hemoglobin solubility in the cytosol. Therefore, the hydropho-
bic valine is also extended outward, but instead of binding
water it seeks a hydrophobic niche with which to bind. When
generally asymptomatic. Using Hb S and Hb C as examples, Hb S is fully oxygenated, the quaternary structure of the
Figure 26-2 illustrates the inheritance of abnormal hemoglo- molecule does not produce a hydrophobic pocket for valine to
bins involving mutations in the gene. bind to, which allows the hemoglobin molecules to remain
soluble in the erythrocyte cytosol like Hb A and maintains the
Incidence normal biconcave disc shape of the RBCs. However, the natural
The highest frequency of the sickle cell trait (Hb AS) is in Africa, allosteric change that occurs upon deoxygenation creates a
where the frequency is 20% to 40%.8 In the United States, hydrophobic pocket in the area of phenylalanine 85 and
approximately 12% of African Americans have a hemoglobin leucine 88, which allows the valine from an adjacent hemo-
variant. The homozygous state (Hb SS) represents the most globin molecule to bind. This hemoglobin pairing creates an
severe type of hemoglobinopathy, with an estimated incidence orientation that helps other hemoglobin molecules to form
of 1 in 375 live African American births.9 The rate of occurrence amino acid bonds and become the seed for polymer formation.
of heterozygous sickle cell trait is 8% among African Ameri- Other hemoglobin pairs polymerize, forming a hemoglobin
cans, with the serious compound heterozygous states of Hb SC core composed of four hemoglobin molecules that elongate in
and Hb S-thal occurring at rates of 1 in 835 (0.12%) and 1 a helical formation. An outer layer of 10 hemoglobin mole-
in 1667 (0.06%), respectively.8 Although in the United States cules forms around the four-hemoglobin-molecule core, creat-
SCD is found mostly in individuals of African descent, it also ing the long, slender Hb S polymer.12-15 Hb S molecules within
has been found in individuals from the Middle East, India, and the RBCs become less soluble, forming tactoids or liquid crys-
the Mediterranean area (Figure 26-3). SCD can also be found tals of Hb S polymers that grow in length beyond the diameter
in individuals from the Caribbean and Central and South of the RBC causing sickling. In homozygotes, the sickling
America.10 The sickle cell mutation is becoming more promi- process begins when oxygen saturation decreases to less than
nent in southern India, particularly in certain tribes.11 85%. In heterozygotes, sickling does not occur unless the
372 PART IV Hematopathology: Erythrocyte Disorders

Thalassemia

Sickle cell anemia

Hb C

Hb D

Hb E

Figure 26-3 Geographic distribution of common inherited structural hemoglobin (Hb) variants and the thalassemias. (From Hoffbrand AV, Pettit JE: Essential
haematology, ed 3, Oxford, 1993, Blackwell Scientific.)

oxygen saturation of hemoglobin is reduced to less than 40%.16 prevents them from entering the microcirculation and causing
The blood becomes more viscous when polymers are formed vasoocclusion.
and sickle cells are created.16 Increased blood viscosity and Not only the oxygen tension but also the level of intra
sickle cell formation slow blood flow. In addition to a decrease cellular hydration affects the sickling process. When RBCs
in oxygen tension, there is a reduction in the pH and an containing Hb S are exposed to a low oxygen tension, hemo-
increase in 2,3-biphosphoglycerate. Reduced blood flow pro- globin polymerization occurs. Polymerized deoxyhemoglobin
longs the exposure of Hb Scontaining erythrocytes to a S activates a membrane channel called Psickle that is otherwise
hypoxic environment, and the lower tissue pH decreases the inactive in normal RBCs. These membrane channels open when
oxygen affinity, which further promotes sickling. The end result the blood partial pressure of oxygen decreases to less than
is occlusion of capillaries and arterioles by sickled RBCs and 50 mm Hg. Open Psickle channels allow the influx of Ca2+, raising
infarction of surrounding tissue. the intracellular calcium levels and activating a second mem-
Sickle cells occur in two forms: reversible sickle cells and brane channel called the Gardos channel. An activated Gardos
irreversible sickle cells.17 Reversible sickle cells are Hb channel causes the efflux of K+, which stimulates the efflux of
Scontaining erythrocytes that change shape in response to Cl through another membrane channel to maintain charge
oxygen tension. Reversible sickle cells circulate as normal equilibrium across the RBC membrane. The efflux of these ions
biconcave discs when fully oxygenated but undergo hemoglo- leads to water efflux and intracellular dehydration, effectively
bin polymerization, show increased viscosity, and change increasing the intracellular concentration of Hb S and intensify-
shape on deoxygenation. The vasoocclusive complications of ing polymerization. Another contributor to K+ and Cl efflux
SCD are thought to be due to reversible sickle cells that are and the resultant dehydration is the K+/Cl cotransporter
able to travel into the microvasculature in the biconcave disk system. Ironically, this system is activated by dehydration and
conformation due to their normal rheologic properties when positively charged hemoglobins such as Hb S and Hb C. The
oxygenated and then become distorted and viscous as they K+/Cl cotransporter pathway is also activated by the low pH
become deoxygenated, converting to the sickle cell configura- encountered in the spleen and kidneys. One potential explana-
tion in the vessel. tion for the altered function of the membrane channels is
In contrast, irreversible sickle cells do not change their shape oxidative damage triggered by Hb S polymerization. Injury to
regardless of the change in oxygen tension or degree of hemo- the RBC membrane induces adherence to endothelial surfaces,
globin polymerization. These cells are seen on the peripheral which causes RBC aggregation, produces ischemia, and exacer-
blood film as elongated sickle cells with a point at each end. bates Hb S polymerization.7
It is thought that irreversible sickle cells are recognized as Another important factor in the pathophysiology of SCD
abnormal by the spleen and removed from circulation, which involves the redistribution of phospholipids in the RBC
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 373

membrane, which contributes to hemolysis, vasoocclusive Clinical Features


crisis, stroke, and acute chest syndrome. In the membranes of The clinical manifestations of SCD can vary from no symptoms
normal RBCs, choline phospholipids like sphingomyelin and to a potentially lethal state with the variety of symptoms listed
phosphatidylcholine are located on the outer surface, whereas in Box 26-3. Hundreds of hemoglobin variants are known;
aminophospholipids like phosphatidylserine (PS) and phos- however, only four are clinically significant: Hb SS, Hb SC, Hb
phatidylethanolamine are primarily on the inner portion of S0-thal, and Hb S+-thal. These four clinically severe forms
the membrane. This asymmetrical distribution of membrane have high morbidity and mortality rates. Average life span for
phospholipids is accomplished by adenosine triphosphate Hb SS patients is 45 years; for Hb SC patients, it is 65 years,
dependent enzymes called translocases or flippases. Inhibition of but life expectancy is increasing over time because of research
flippases and activation of an enzyme called scramblase cause a findings and improved patient care.27 Individuals affected with
more random distribution of membrane phospholipids, which SCD are characteristically symptom free until the second half
increases the number of choline phospholipids on the interior of the first year of life because of the protective effect of Hb F.28
half of the membrane and the number of aminophospholipids Toward the end of the first 6 months of life, mutated chains
on the exterior membrane surface. The sickle cells of homozy- begin to be produced and gradually replace normal chains,
gotes (Hb SS) express 2.1% PS on erythrocyte exterior surfaces which causes Hb S levels to increase and Hb F levels to decrease.
compared with 0.2% for normal Hb AA controls.18,19 It is Erythrocytes containing Hb S become susceptible to hemolysis,
hypothesized that Hb S polymerization may produce micro and a progressive hemolytic anemia and splenomegaly may
particles and iron complexes that adhere to the RBC membrane become evident.
and generate reactive oxygen species, which, along with Many individuals with SCD undergo episodes of recurring
increased intracellular calcium or protein kinase C activation, pain termed crises. Sickle cell crises were described by Diggs as
may contribute to flippase inhibition and scramblase activa- any new syndrome that develops rapidly in patients with SCD
tion.20,21 PS on the exterior surface of RBCs binds thrombos- owing to the inherited abnormality.29 The pathogenesis of
pondin on vascular endothelial cells,22 enhancing adherence the acute painful episode first described by Diggs is not
between RBCs and the vessel wall and contributing to vasooc- fully understood. Various crises may occur: vasoocclusive or
clusive crisis, activation of coagulation, and decreased RBC painful, aplastic, megaloblastic, sequestration, and chronic
survival.23,24 In addition, RBCs with PS on the external mem- hemolytic.
brane surface are vulnerable to hydrolysis by secretory phos- The hallmark of SCD is vasoocclusion, which accounts for
pholipase A2 (sPLA2), which generates lysophospholipids and most hospital and emergency department visits. This acute,
fatty acids like lysophosphatidic acid. This results in vascular painful aspect of SCD occurs with great predictability and
damage that contributes to acute chest syndrome.25,26 severity in many individuals and can be triggered by acidosis,

BOX 26-3 Clinical Features of Sickle Cell Disease


I. Vasoocclusion 5. Penis
A. Causes Priapism
Acidosis 6. Eyes
Hypoxia Retinal hemorrhage
Dehydration 7. Central nervous system
Infection 8. Urinary tract
Fever Renal papillary necrosis
Extreme cold 9. Leg ulcers
B. Clinical manifestations II. Bacterial infections
1. Bones A. Sepsis
Pain B. Pneumonia
Hand-foot dactylitis C. Osteomyelitis
Infection (osteomyelitis) III. Hematologic defects
2. Lungs A. Chronic hemolytic anemia
Pneumonia B. Megaloblastic episodes
Acute chest syndrome C. Aplastic episodes
3. Liver IV. Cardiac defects
Hepatomegaly A. Enlarged heart
Jaundice B. Heart murmurs
4. Spleen V. Other clinical features
Sequestration splenomegaly A. Stunted growth
Autosplenectomy B. High-risk pregnancy
374 PART IV Hematopathology: Erythrocyte Disorders

hypoxia, dehydration, infection and fever, and exposure to slow blood flow.32 Cells of patients with Hb SC disease produce
extreme cold. Painful episodes manifest most often in the less sickling with fewer adherent RBCs.5,33
bones, lungs, liver, spleen, penis, eyes, central nervous system, The frequency of painful episodes varies from none to six
and urinary tract. per year.5 On average, each episode persists for 4 to 5 days,
The pathogenesis of vasoocclusion in SCD is not fully although protracted episodes may last for weeks. Repeated
understood, but Hb S polymerization and sickling of RBCs splenic infarcts produce scarring, resulting in diminished
play a major role, with other factors also affecting this process. splenic tissue and abnormal function. Splenic sequestration is
The list of possible risk factors includes polymerization, characterized by a sudden trapping of blood in the spleen,
decreased deformability, sickle cellendothelial cell adherence, which leads to a rapid decline in hemoglobin, often to less
endothelial cell activation, white blood cell (WBC) and platelet than 6 g/dL.33 This phenomenon occurs most often in infants
activation, hemostatic activation, and vascular tone.29 The and young children whose spleens are chronically enlarged.
interrelationships among these risk factors is shown in Figure Children experiencing splenic sequestration episodes may have
26-4. Vasoocclusion can be triggered by any of these factors earlier onset of splenomegaly and a lower level of Hb F at 6
under various circumstances. During inflammation, increased months of age.5 Crises are often associated with respiratory
WBCs interacting with endothelium, platelet activation causing tract infections. Gradual loss of splenic function is referred to
elevation of thrombospondin level, or clinical dehydration as autosplenectomy and is evidenced by the presence of Howell-
resulting in an increase in von Willebrand factor can trigger Jolly and Pappenheimer bodies on the peripheral blood film.
RBC adherence to endothelium, precipitating vascular obstruc- In the lungs, pulmonary infarction from sickling in the
tion. Another mechanism of obstruction can be dense cells, microvasculature causes acute chest syndrome.
which are less deformable and are at greatest risk for intracel- Acute chest syndrome is characterized by fever, chest pain,
lular polymerization because of their higher Hb S concentra- and presence of pulmonary infiltrates on the chest radiograph,
tion.30,31 Vasoocclusive episodes gradually consume the patient and is the leading cause of death among adults with SCD. Over
organ by organ, through the destructive and debilitative effects 10% of adults with acute chest syndrome die from com
of cumulative infarcts. Approximately 8% to 10% of SCD plications linked to chronic lung disease and pulmonary
patients develop cutaneous manifestations in the form of hypertension.34 In children, acute chest syndrome generally is
ulcers or sores on the lower leg.8 precipitated by infection characterized by fever, cough, and
The abnormal interaction between sickle cells and vascular tachypnea. Acute chest syndrome is also linked with sPLA2,
endothelium seems to have a great impact on the vasoocclusive discussed previously. The level of sPLA2 has been shown to be
event. Endothelial adherence correlates significantly with the a predictor of acute chest syndrome in patients with SCD35 in
severity of painful episodes. In addition, sickle cell adherence that sPLA2 rises 24 to 48 hours before symptoms of acute chest
to vascular endothelium results in intimal hyperplasia that can syndrome begin.36 In addition, a high sPLA2 level correlates
with the degree of lung damage.
Pulmonary hypertension (PHT) is a serious and potentially
fatal sequela of SCD. Among patients with SCD, PHT has a
INFECTION/INFLAMMATION C-reactive protein prevalence of about 33%, with 10% of patients manifesting
Tissue factor expression a more severe version. The mortality rate for sickle cell patients
Release of biologic modifiers who develop PHT is 40% at 40 months. An association has
COAGULATION ACTIVATION been documented between the development of PHT and the
WBC ACTIVATION nitrous oxide (NO) pathway. NO is produced from the action
Thrombin generation
ENDOTHELIAL MODULATION
of endothelial NO synthase (eNOS) on arginine, which causes
PLATELET ACTIVATION
vasodilation. Patients with SCD have a decrease in NO, and
this leads to vasoconstriction and hypertension.1,37 In addi-
RBC ADHESION TO ENDOTHELIUM Thrombospondin release tion, low NO levels in the blood fail to inhibit endothelin-1,
a potent vasoconstrictor, which results in additional vasocon-
VASCULAR OBSTRUCTION
striction and hypertension.34 The connection between NO and
von Willebrand factor release
SCD involves the hemolytic crisis. Erythrocyte hemolysis
POOR RBC DEFORMABILITY releases high levels of arginase, which degrades arginine; the
Altered vascular tone
Acidosis
result is less NO production from eNOS.38,39 In addition, the
CLINICAL DEHYDRATION
free hemoglobin released from hemolyzed RBCs scavenges
RBC dehydration RBC SICKLING HYPOXIA NO, which further reduces the levels and exacerbates the
vasoconstriction and hypertension.38 Blood arginine and NO
Delayed blood flow levels drop a few days before the onset of acute chest
Figure 26-4 Numerous risk factors for vasoocclusion are highly interre- syndrome,33 a finding suggesting that the NO pathway is a
lated physiologically, as shown here. RBC, Red blood cell; WBC, white blood connection between SCD, PHT, and asthma.34,40 Treatment
cell. (From Embury SH, Hebbel RP, Mohandas N, etal: Sickle cell disease: with large doses of arginine reduces pulmonary artery pres-
basic principles and clinical practice, Philadelphia, 1994, Lippincott Williams sure, but the effect is not sustainable and does not reduce
& Wilkins, p 322.) mortality.
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 375
Bacterial infections pose a major problem for SCD patients. Doppler ultrasonography or magnetic resonance angiography
These patients have increased susceptibility to life-threatening is recommended to detect microstrokes.42
infection from Staphylococcus aureus, Streptococcus pneumoniae,
and Haemophilus influenzae. Acute infections are common Incidence with Malaria and Glucose-6-Phosphate
causes of hospitalization and have been the most frequent Dehydrogenase Deficiency
cause of death, especially in the first 3 years of life.1 Bacterial The sickle gene occurs with greatest frequency in Central Africa,
infections of the blood (septicemia) are exacerbated by the the Near East, the region around the Mediterranean, and parts
autosplenectomy effect as the spleen gradually loses its ability of India. The frequency of the gene parallels the incidence of
to function as a secondary lymphoid tissue to effectively clear P. falciparum and seems to offer some protection against cere-
organisms from the blood. bral falciparum malaria in young patients. Malarial parasites
Chronic hemolytic anemia is characterized by shortened are living organisms within the RBCs that use the oxygen
RBC survival with a corresponding decrease in hemoglobin within the cells. This reduced oxygen tension causes the cells
and hematocrit (Hct), an elevated reticulocyte count, and jaun- to sickle, which results in injury to the cells. These injured cells
dice. Continuous screening and removal of sickle cells by the tend to become trapped within the blood vessels of the spleen
spleen perpetuate the chronic hemolytic anemia and autosple- and other organs, where they are easily phagocytized by scav-
nectomy effect. Because other conditions, such as hepatitis and enger WBCs. Selective destruction of RBCs containing parasites
gallstones, may cause jaundice, chronic hemolysis is difficult decreases the number of malarial organisms and increases the
to diagnose in sickle cell patients.17 time for immunity to develop. One explanation for this
Megaloblastic episodes result from the sudden arrest of phenomenon is that the infected cell is uniquely sickled and
erythropoiesis due to folate depletion. Folic acid deficiency as destroyed, probably in an area of the spleen or liver where
a cause of exaggerated anemia in SCD is extremely rare in the phagocytic cells are plentiful, and the oxygen tension is
United States. It is common practice to prescribe prophylactic significantly decreased.44
folic acid for patients with SCD, however.5 Because of the high incidence of glucose-6-phosphate dehy-
Aplastic episodes (bone marrow failure) are the most drogenase (G6PD) deficiency in patients with SCD, it has been
common life-threatening hematologic complications and are suggested that G6PD deficiency has a protective effect in these
usually associated with infection, particularly parvovirus infec- patients,45 although this correlation has not been confirmed
tion.31 Aplastic episodes present clinical problems similar to through studies. It also has been postulated that hemolytic
those seen with other hemolytic disorders.41 Sickle cell patients episodes are more common in these patients, but research has
usually can compensate for the decrease in RBC survival by shown no relationship between the clinical severity of sickling
increasing bone marrow output. When the bone marrow is and the presence or absence of G6PD deficiency. Because of
suppressed temporarily by bacterial or viral infections, however, the presence of young cells rich in G6PD, however, the
the hematocrit decreases substantially with no reticulocyte increased hemolysis is more likely caused by the enzyme
compensation. The spontaneous recovery phase is character- abnormality when the population is shifted to the oldest cells
ized by the presence of nucleated RBCs and an increase in the during an aplastic crisis.46
number of reticulocytes in the peripheral blood. Most aplastic
episodes are short-lived and require no therapy. If anemia is Laboratory Diagnosis
severe and the bone marrow remains aplastic, transfusions The anemia of SCD is a chronic hemolytic anemia, classified
become necessary. If patients are not transfused in a timely morphologically as normocytic, normochromic. The character-
fashion, death can occur.41 istic diagnostic cell observed on a Wright-stained peripheral
Patients also experience cardiac defects, including enlarged blood film is a long, curved cell with a point at each end
heart and heart murmurs. In patients with severe anemia, car- (Figure 26-5). Because of its appearance, the cell was named a
diomegaly can develop as the heart works to maintain high sickle cell.29 The peripheral blood film shows marked poikilo-
blood flow. Increased cardiac workload along with increased cytosis and anisocytosis with normal RBCs, sickle cells, target
bone marrow erythropoiesis increases calorie burning, con cells, nucleated RBCs along with a few spherocytes, basophilic
tributing to a reduced growth rate.42 When patients enter stippling, Pappenheimer bodies, and Howell-Jolly bodies. The
childbearing age, pregnancy becomes risky.1 presence of sickle cells and target cells is the hallmark of SCD.
Impaired blood supply to the head of the femur and There is moderate to marked polychromasia with a reticulocyte
humerus results in a condition called avascular necrosis (AVN). count between 10% and 25%, corresponding with the hemo-
About 50% of patients with SCD develop AVN by 35 years of lytic state and the resultant bone marrow response. The RBC
age.43 Physical therapy and surgery to relieve intramedullary distribution width (RDW) is increased owing to moderate
pressure within the head of the long bones are effective, but anisocytosis. The mean cell volume (MCV) is not as elevated
hip and/or shoulder implants become necessary in most as one would expect, however, given the elevated reticulocyte
patients experiencing AVN.43 count. An aplastic crisis can be heralded by a decreased
Microstrokes can lead to headaches, poor school perfor- reticulocyte count. Moderate leukocytosis is usually present
mance, reduced intelligence quotient (IQ), and overt central (sometimes 40 to 50 109 WBCs/L) with neutrophilia and a
nervous system dysfunction. A neurologic examination followed mild shift toward immature granulocytes. Thrombocytosis is
by magnetic resonance imaging and, if available, transcranial usually present. The leukocyte alkaline phosphatase score is
376 PART IV Hematopathology: Erythrocyte Disorders

A B
Figure 26-5 A, Peripheral blood film for a patient with sickle cell disease (SCD) showing anisocytosis, polychromasia, three sickle cells, target cells, and
normal platelets (1000). B, Peripheral blood film for an SCD patient showing anisocytosis, poikilocytosis, sickle cells, target cells, and one nucleated RBC
(1000). Platelets are not present in this field, but their numbers were adequate in this patient. (Courtesy Ann Bell, University of Tennessee, Memphis.)

not elevated when neutrophilia is caused by sickle cell crisis


alone, and no underlying infection is present. The bone marrow
shows erythroid hyperplasia reflecting an attempt to compen-
sate for the anemia, which results in polychromasia and an
increase in reticulocytes and nucleated RBCs in the peripheral
blood. Levels of immunoglobulins, particularly immuno
globulin A, are elevated in all forms of SCD. Serum ferritin
levels are normal in young patients but tend to be elevated later
in life; however, hemochromatosis is rare. Chronic hemolysis
is evidenced by elevated levels of indirect and total bilirubin
with the accompanying jaundice.
The diagnosis of SCD is made by demonstrating the insolu-
bility of Hb S in the deoxygenated form using a screening test,
followed by confirmation of its presence using hemoglobin
electrophoresis, high-performance liquid chromatography
(HPLC), or capillary electrophoresis. An older screening test
detects Hb S insolubility by inducing sickle cell formation on
a glass slide. A drop of blood is mixed with a drop of 2%
sodium metabisulfite (a reducing agent) on a slide, and the
mixture is sealed under a coverslip. The hemoglobin inside the
RBCs is reduced to the deoxygenated form; this induces poly
merization and the resultant sickle cell formation, which can
be identified microscopically. This method is slow and Figure 26-6 Tube solubility screening test for the presence of hemoglobin
cumbersome and is rarely used. S. In a negative test result (left ), the solution is clear and the lines behind
The decreased solubility of deoxygenated Hb S in solution the tube are visible. In a positive test result (right ), the solution is turbid and
forms the basis for the hemoglobin solubility test, the most the lines are not visible. (Courtesy Ann Bell, University of Tennessee,
common screening test. Blood is added to a buffered salt solu- Memphis.)
tion containing a reducing agent, such as sodium dithionite,
and a detergent-based lysing agent (saponin). The saponin
causes the release of the hemoglobin from the RBCs, and the
dithionite converts the hemoglobin to the deoxygenated form. that give a positive result on the solubility test include Hb
Deoxygenated Hb S is insoluble and precipitates in solution, C-Harlem (Georgetown), Hb C-Ziguinchor, Hb S-Memphis,
which renders it turbid, whereas solutions containing nonsick- Hb S-Travis, Hb S-Antilles, Hb S-Providence, Hb S-Oman, Hb
ling hemoglobins remain clear (Figure 26-6). False-positive Alexander, and Hb Porte-Alegre.1,5 All of these hemoglobins
results for Hb S can occur with hyperlipidemia, a few rare have two amino acid substitutions, the Hb S substitution
hemoglobinopathies, and heavily inoculated test solutions; (6GluVal) and another unrelated substitution. Hb S-Antilles
false-negative results can occur because of an inadequate is particularly important because it can cause sickling in the
number of RBCs or a low hematocrit. Other hemoglobins heterozygous state.
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 377
Alkaline hemoglobin electrophoresis is a common first step American Pathologists survey, however, hemoglobin electro-
in the definitive diagnosis of hemoglobinopathies, including phoresis in agarose medium is still the most commonly used
SCD. Over the years hemoglobin electrophoresis on cellulose technique to identify hemoglobin variants.47 The more progres-
acetate medium has been replaced by electrophoresis on sive laboratories use a combination of two or more techniques
agarose medium. Nonetheless, because some hemoglobins to improve identification of hemoglobin variants. Some refer-
have the same electrophoretic mobility patterns, hemoglobins ence laboratories may use isoelectric focusing to separate
that exhibit an abnormal electrophoretic pattern at an alkaline hemoglobin types based on isoelectric point.48
pH may be subjected to electrophoresis at an acid pH for Patients with Hb SS or Hb SC disease lack normal -globin
definitive separation. For example, Hb S migrates with Hb D chains, so they have no Hb A. Hb S level is usually greater than
and Hb G on alkaline electrophoresis but separates from Hb 80%. The Hb F level is usually increased (1% to 20%), and
D and Hb G on acid electrophoresis. Similarly, Hb C migrates when Hb F constitutes more than 20% of hemoglobin, it has
with Hb E and Hb O on alkaline electrophoresis but separates a tendency to modulate the severity of the disease. This is
on acid electrophoresis. Figure 26-7 shows electrophoretic pat- especially true in newborns and in patients with hereditary
terns for abnormal hemoglobins. HPLC and capillary electro- persistence of fetal hemoglobin.28 The Hb A2 level is normal or
phoresis are gaining in popularity, because these methods are slightly increased (2% to 5%), and Hb A2 quantitation is useful
more automated and the instruments more user friendly. in differentiating SCD from Hb S0-thalassemia, in which Hb
HPLC separates hemoglobin types in a cation exchange column A2 is increased (see Chapter 27). Hb G, Hb D, and Hb S all
and usually requires only one sample injection. Unlike electro- migrate in the same position on alkaline cellulose acetate and
phoresis, HPLC can identify and quantitate low levels of Hb alkaline agarose electrophoresis, but Hb G and Hb D do not
A2 and Hb F, but co-migration of Hb A2 and Hb E occurs. give a positive result on the tube solubility test.
Capillary electrophoresis separates hemoglobin types based on The typical sequelae of SCD may be predicted, and the
charge in an alkaline buffer, but does so using smaller volumes effectiveness of treatment monitored, if reliable biomarkers of
and produces better separation than traditional agarose elec- inflammation can be identified. Among the common indica-
trophoresis. According to data from a 2007 to 2009 College of tors of inflammation, WBC is a good predictor of sickle cell
events and mortality, whereas the erythrocyte sedimentation
rate and C-reactive protein (CRP) level exhibit variability too
great to reliably predict events. However, CRP and sPLA2 are
both elevated during vasoocclusive crisis and acute chest syn-
drome. Other markers such as interleukin-6 (IL-6), IL-10, and
protein S are showing promise as useful indicators in clinical
practice.49 Lipid damage from oxidative stress can be predicted
by plasma elevations of malondialdehyde (MDA) and depleted
-tocopherol. In addition, -tocopherol rises with CRP during
bouts of inflammation. Of all the biomarkers evaluated, IL-6,
IL-10, vascular cell adhesion molecule 1 (VCAM-1), and sPLA2
are the most promising at predicting impending crisis.50

Treatment
Supportive care has been the mainstay of therapy for SCD. New
therapies have evolved, however, that are actually modifying
the genetic pathogenesis of the disease. Neonatal screening,
childhood prophylactic penicillin therapy, bone marrow trans-
plantation, and treatment with hydroxyurea in adults may
extend the life of the SCD patient further.
The main components of supportive therapy include ade-
quate hydration, prophylactic vitamin therapy, avoidance of
low-oxygen environments, analgesia for pain, and aggressive
antibiotic therapy with the first signs of infection. Hydration
maintains good blood flow and reduces vasoocclusive crises.
Prophylactic oral penicillin V at a dose of 125 mg twice per day
Figure 26-7 Relative mobilities of normal and variant hemoglobins (Hb)
by the age of 3 months is recommended to avoid infection and
in various conditions measured by electrophoresis on cellulose acetate at an
the associated morbidity and mortality. The penicillin dosage
alkaline pH and citrate agar at an acid pH. The relative amount of hemoglobin
is not proportional to the size of the band; for example, in sickle cell trait (Hb is increased to 250 mg twice per day at 3 years of age.51 When
AS), the bands may appear equal, but the amount of Hb A exceeds that of infections occur, prompt antibiotic treatment reduces the
Hb S. (From Schmidt RM, Brosious EF: Basic laboratory methods of hemo- associated morbidity and mortality.6 Avoidance of strenuous
globinopathy detection, ed 6, HEW Pub No (CDC) 77-8266, Atlanta, 1976, exercise, high altitudes, and unpressurized air travel maintains
Centers for Disease Control and Prevention.) high oxygen tensions and reduces the sickling phenomenon.
378 PART IV Hematopathology: Erythrocyte Disorders

Treatment for painful episodes includes ensuring optimal survival rates for patients receiving transplants from HLA-
hydration, rapidly treating associated infection, and effectively identical related donors are between 80% and 90% for
relieving pain. Analgesics are the foundation of pain manage- SCD.58-61 Patients chosen for transplantation are generally chil-
ment, with nonsteroidal antiinflammatory drugs administered dren younger than age 17 with severe complications of SCD
to manage mild ischemic attacks. Opioids are recommended (i.e., stroke, acute chest syndrome, and refractory pain). In
when pain becomes chronic.52 The patient should be examined addition, morbidity and mortality following transplantation
on a regular basis, and routine testing should be done to estab- increases with age, which places another restriction on trans-
lish normal values for the patient during nonsickling periods. plantation therapy.62 There is evidence that transplantation
Children younger than 3 years often experience hand and restores some splenic function, but its effect on established
foot syndrome, characterized by pain and swelling in the hands organ damage is unknown.63 Transplantation of cord blood
and feet.30 Treatment usually consists of increasing intake of stem cells from HLA-identical related and unrelated donors
fluids and giving analgesics for pain. is associated with a disease-free survival rate of 90%. The
Pneumococcal disease has been a leading cause of morbid- primary benefit of using cord blood as a source of stem cells
ity and mortality in children, especially children younger than is that banking of cord blood increases the number of
6 years. With immunization and prophylactic antibiotics, units available to achieve an HLA match.64 Some researchers
however, this is now a preventable complication.4 Immuni are now focusing on the use of in utero stem cell transplanta-
zation with heptavalent conjugated pneumococcal vaccine is tion to produce engraftment while the immune system of
recommended at 2, 4, and 6 months of age. The 23-valent the fetus is prone to HLA tolerance. Others are attempting to
pneumococcal vaccine is recommended at 2 years with a genetically alter fetal hematopoietic stems cells to overcome
booster at the age of 5 years. Standard childhood vaccinations HLA mismatches.65
should be given as scheduled. In addition, annual administra- Hydroxyurea therapy has offered some promise in relieving
tion of influenza vaccine is recommended beginning at 6 the sickling disorder by increasing the proportion of Hb F in
months of age.42 The risk of bacterial infection probably the erythrocytes of individuals with SCD.66 Hydroxyurea, given
increases in mature patients with Hb SC disease and homozy- at 25 to 30 mg/kg, has been shown to reduce symptoms and
gous SCD.17 prolong life, in part by increasing Hb F levels. Because Hb F
Transfusions can be used to prevent the complications of does not copolymerize with Hb S, if the production of Hb F
SCD. More specifically, periodic transfusions, given at a fre- can be sufficiently augmented, the complications of SCD might
quency of eight or more per year, are effective at preventing be avoided. The severity of the disease expression and the
stroke, symptomatic anemia, brain injury, priapism, leg ulcers, number of irreversible sickle cells are inversely proportional to
PHT, delayed pubescence, splenomegaly, and chronic pain, the extent to which Hb F synthesis persists. Individuals in
and improving school attendance, IQ, energy, exercise toler- whom Hb F levels stabilize at 12% to 20% of total hemoglobin
ance, mood, and sense of well-being. Nonetheless, transfusion may have little or no anemia and few, if any, vasoocclusive
therapy has the potential to cause transfusion reactions, attacks. Levels of 4% to 5% Hb F may modulate the disease,
transfusion-related infections, and iron overload. Of the three, and levels of 5% to 12% may suppress the severity of hemolysis
iron overload is the most frequent. Iron overload has been and lessen the frequency of severe episodes.31
associated with endocrine dysfunction53 and cardiac disease.54
Deferoxamine has been effective in treating iron overload by Course and Prognosis
chelating and removing much of the excess iron from the body. Proper management of SCD has increased the life expectancy
Deferoxamine must be administered intravenously, however, of patients from 14 years in 1973 to the current average life
and treatment requires at least 8 hours each day for a week. A span of 50 years.67 For men and women who are compound
new oral iron chelator, deferasirox, was approved by the Food heterozygotes for Hb SC, the average life span is 60 and 68
and Drug Administration in 2005.55 Deferasirox is consumed years, with a few patients living into their seventies.27,68 Indi-
in the morning as a slurry by dissolving several pills, but its viduals with Hb SS can pursue a wide range of vocations and
effectiveness is yet to be determined. In other circumstances, professions. They are discouraged, however, from jobs that
such as central nervous system infarction, hypoxia with infec- require strenuous physical exertion or exposure to high alti-
tion, stroke, episodes of acute chest syndrome, and preparation tudes or extreme environmental temperature variations.
for surgery, transfusions are used to decrease blood viscosity Newborn screening for hemoglobinopathies has signifi-
and the percentage of circulating sickle cells. Before surgery, cantly reduced mortality in children with SCD by enabling
Hb SS patients are transfused with normal Hb AA blood to prompt and comprehensive medical care. The most common
bring the volume of Hb S to less than 50% in an effort to form of screening is HPLC followed by confirmation using
prevent complications in surgery.56 Maintenance transfusions hemoglobin electrophoresis and genotyping methods.69
should be given in pregnancy if the mother experiences vasooc-
clusive or anemia-related problems or if there are signs of fetal Sickle Cell Trait
distress or poor growth.17 The term sickle cell trait refers to the heterozygous state (Hb
Bone marrow or stem cell transplantation has proved suc- AS) and describes a benign condition that generally does not
cessful for some individuals, but few patients qualify due to affect mortality or morbidity. The trait occurs in approximately
the lack of HLA-matched, related donors.57 The event-free 8% of African Americans. It also can be found in Central
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 379

Americans, Asians, and people from the region around the


Mediterranean.1
Individuals with sickle cell trait are generally asymptomatic
and present with no significant clinical or hematologic mani-
festations. Under extremely hypoxic conditions, however, sys-
temic sickling and vascular occlusion with pooling of sickled
cells in the spleen, focal necrosis in the brain, and even death
can occur. In circumstances such as severe respiratory infection,
unpressurized flight at high altitudes, and anesthesia in which
pH and oxygen levels are sufficiently lowered to cause sickling,
patients may develop splenic infarcts.5 Failure to concentrate
urine is the only consistent abnormality found in these
patients.70 This abnormality is caused by diminished perfusion
of the vasa recta of the kidney, which impairs concentration of
urine by the renal tubules. Renal papillary necrosis with hema- Figure 26-8 Peripheral blood film for a patient with hemoglobin (Hb) C
turia has been described in some patients.5 disease showing one Hb C crystal and target and folded cells (1000).
(Courtesy Ann Bell, University of Tennessee, Memphis.)
The peripheral blood film of a patient with sickle cell trait
shows normal RBC morphology, with the exception of a few
target cells. No abnormalities in the leukocytes and thrombo- Laboratory Diagnosis
cytes are seen. The hemoglobin solubility screening test yields A mild to moderate, normochromic, normocytic anemia occurs
positive results, and sickle cell trait is diagnosed by detecting in homozygous Hb C disease. Occasionally, some microcytosis
the presence of Hb S and Hb A on hemoglobin electrophoresis and mild hypochromia may be present. There is a marked
or HPLC. In individuals with sickle cell trait, electrophoresis increase in the number of target cells, a slight to moderate
reveals approximately 40% or less Hb S and approximately increase in the number of reticulocytes, and nucleated RBCs
60% or more Hb A, Hb A2 level is normal or slightly increased, may be present in the peripheral blood.
and Hb F level is within normal limits. Levels of Hb S less than Hexagonal crystals of Hb C form within the erythrocyte and
40% can be seen in patients who also have -thalassemia or may be seen on the peripheral blood film (Figure 26-8). Many
iron or folate deficiency.17 No treatment is required for this crystals appear extracellularly with no evidence of a cell mem-
benign condition, and the patients life span is not affected by brane.71,72 In some cells, the hemoglobin is concentrated within
sickle cell trait. the boundary of the crystal. The crystals are densely stained and
vary in size and appear oblong with pyramid-shaped or pointed
ends. These crystals may be seen on wet preparations by
HEMOGLOBIN C
washing RBCs and resuspending them in a solution of sodium
Hb C was the next hemoglobinopathy after Hb S to be described citrate.8
and in the United States is found almost exclusively in the Hb C yields a negative result on the hemoglobin solubility
African American population. Spaet and Ranney reported this test, and definitive diagnosis is made using electrophoresis or
disease in the homozygous state (Hb CC) in 1953.5 HPLC. No Hb A is present in Hb CC disease. In addition, Hb
C is present at levels of greater than 90%, with Hb F at less
Incidence, Etiology, and Pathophysiology than 7% and Hb A2 at approximately 2%. In Hb AC trait, about
Hb C is found in 17% to 28% of people of West African 60% Hb A and 30% Hb C are present. On cellulose acetate
extraction and in 2% to 3% of African Americans. It is the electrophoresis at an alkaline pH, Hb C migrates in the same
most common nonsickling variant encountered in the United position as Hb A2, Hb E, and Hb O-Arab (see Figure 26-7). Hb
States and the third most common in the world.1 Hb C is C is separated from these other hemoglobins on citrate agar
defined by the structural formula 226GluLys, in which lysine electrophoresis at an acid pH (see Figure 26-7). No specific
is substituted for glutamic acid in the sixth position of the treatment is required. This disorder becomes problematic only
chain. Lysine has a +1 charge, so the result of this substitu- if infection occurs or if mild chronic hemolysis leads to
tion is a net change in charge of +2, which has a different gallbladder disease.
structural effect on the hemoglobin molecule than the Hb S
substitution.
HEMOGLOBIN C-HARLEM
Hb C is inherited in the same manner as Hb S but manifests
(HEMOGLOBIN C-GEORGETOWN)
as a milder disease. Similar to Hb S, Hb C polymerizes under
low oxygen tension, but the structure of the polymers differs. Hb C-Harlem (also known as Hb C-Georgetown) has a double
Hb S polymers are long and thin, whereas the polymers in Hb substitution on the chain.17,73 The substitution of valine for
C form a short, thick crystal within the RBCs. The shorter Hb glutamic acid at the sixth position of the chain is identical
C crystal does not alter RBC shape to the extent that Hb S does, to the Hb S substitution, and the substitution at the seventy-
so there is less splenic sequestration and hemolysis. In addi- third position of aspartic acid for asparagine is the same as that
tion, vasoocclusive crisis does not occur. in the Korle Bu mutation. The double mutation is termed Hb
380 PART IV Hematopathology: Erythrocyte Disorders

C-Harlem (Hb C-Georgetown) because the abnormal hemoglo- protection against malaria.1 Hb E trait is asymptomatic. When
bin migrates with Hb C on cellulose acetate electrophoresis at Hb E is combined with -thalassemia, however, the disease
an alkaline pH. Patients with this anomaly are asymptomatic, becomes more severe than Hb EE and more closely resembles
but patients with compound heterozygosity for Hb S and Hb -thalassemia major, requiring regular blood transfusions.1
C-Harlem have crises similar to those in Hb SS disease.
A positive solubility test result may occur with Hb C-Harlem, Laboratory Diagnosis
and hemoglobin electrophoresis or HPLC is necessary to Hb E does not produce a positive hemoglobin solubility test
confirm the diagnosis. On cellulose acetate at pH 8.4, Hb result and must be confirmed using electrophoresis or HPLC.
C-Harlem migrates in the C position (see Figure 26-7). Citrate In the homozygous state there is greater than 90% Hb E, a very
agar electrophoresis at pH 6.2, however, shows migration of low MCV (55 to 65 fL), few to many target cells, and a normal
Hb C-Harlem in the S position (see Figure 26-7). Because so reticulocyte count. The heterozygous state has a mean MCV of
few cases have been identified, the clinical outcome for indi- 65 fL, slight erythrocytosis, target cells,1 and approximately
viduals affected with this abnormality is uncertain.73 30% to 40% Hb E. On cellulose acetate electrophoresis at an
alkaline pH, Hb E migrates with Hb C, Hb O, and Hb A2 (see
Figure 26-7). On citrate agar electrophoresis at an acid pH, Hb
HEMOGLOBIN E
E can be separated from Hb C, but it co-migrates with Hb A
Incidence, Etiology, and Pathophysiology and Hb O (see Figure 26-7).
Hb E was first described in 1954.73,74 The variant occurs with
an incidence of 30% in Southeast Asia. As a result of the influx Treatment and Prognosis
of immigrants from this area, Hb E prevalence has increased No therapy is required with Hb E disease and trait. Some
in the United States.75 It occurs infrequently in African Ameri- patients may experience splenomegaly and fatigue, however.
cans and whites. Hb E is a chain variant in which lysine is Genetic counseling is recommended, and the Hb E gene should
substituted for glutamic acid in the twenty-sixth position be discussed in the same way as a mild thalassemia allele.76
(2226GluLys). As with Hb C, this substitution results in a net
change in charge of +2, but because of the position of the
HEMOGLOBIN O-ARAB
substitution, hemoglobin polymerization does not occur.
Hb O-Arab is a chain variant caused by the substitution
Clinical Features of lysine for glutamic acid at the 121st amino acid position
The homozygous state (Hb EE) manifests as a mild anemia (22121GluLys).5,17,29 It is a rare disorder found in Kenya, Israel,
with microcytes and target cells (Figure 26-9). The RBC survival Egypt, and Bulgaria, and in 0.4% of African Americans. No
time is shortened. The condition is not associated with clini- clinical symptoms are exhibited by individuals who carry this
cally observable icterus, hemolysis, or splenomegaly. The main variant, except for a mild splenomegaly in homozygotes. When
concern in identifying homozygous Hb E is differentiating it Hb O-Arab is inherited with Hb S, however, severe clinical
from iron deficiency, -thalassemia trait, and Hb E-thal (see conditions similar to those in Hb SS result.
Chapter 27).76 The disease, Hb EE, resembles thalassemia trait. Homozygous individuals have a mild hemolytic anemia
Because the highest incidence of the Hb E gene is in the areas with many target cells on the peripheral blood film and a nega-
of Thailand where malaria is most prevalent, it is thought that tive result on the hemoglobin solubility test. The presence of
P. falciparum multiplies more slowly in Hb EE RBCs than in this hemoglobin variant must be confirmed using electropho-
Hb AE or Hb AA RBCs and the mutation may give some resis or HPLC. Because Hb O-Arab migrates with Hb A2, Hb C,
and Hb E on cellulose acetate at an alkaline pH, citrate agar
electrophoresis at an acid pH is required to differentiate it from
Hb C (see Figure 26-7). Hb O-Arab is the only hemoglobin to
move just slightly away from the point of application toward
the cathode on citrate agar at an acid pH. No treatment is
generally necessary for individuals with Hb O-Arab.

HEMOGLOBIN D AND HEMOGLOBIN G


Hb D and Hb G are a group of at least 16 chain variants (Hb
D) and 6 chain variants (Hb G) that migrate in an alkaline
pH at the same electrophoretic position as Hb S.4-6,72 This is
because their and subunits have one less negative charge
at an alkaline pH than Hb A, as does Hb S. They do not sickle,
however, when exposed to reduced oxygen tension.
Figure 26-9 Microcytes and target cells in a patient with hemoglobin E Most variants are named for the place where they were
trait. (From Hematology tech sample H-1, Chicago, 1991, American Society discovered. Hb D-Punjab and Hb D-Los Angeles are identical
of Clinical Pathologists.) hemoglobins in which glutamine is substituted for glutamic
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 381

acid at the 121st position in the chain (22121GluGln). Hb the teenage years. Hb SC disease may cause all the vasoocclu-
D-Punjab occurs in about 3% of the population in northwest- sive complications of sickle cell anemia, but the episodes are
ern India, and Hb D-Los Angeles is seen in fewer than 2% of less frequent, and damage is less disabling. Hemolytic anemia
African Americans. is moderate, and many patients exhibit moderate spleno
Hb G-Philadelphia is an chain variant of the G hemoglo- megaly. Proliferative retinopathy is more common and more
bins with a substitution of asparagine by lysine at the sixty- severe than in sickle cell anemia.82 Respiratory tract infections
eighth position. The Hb G-Philadelphia variant is the most with S. pneumoniae are common.5
common G variant encountered in African Americans and is Patients with Hb SC disease live longer than patients with
seen with greater frequency than the Hb D variants. The Hb G Hb SS and have fewer painful episodes, but this disorder is
variant is also found in Ghana. associated with considerable morbidity and mortality, espe-
Hb D and Hb G do not sickle and yield a negative hemo- cially after age 30.83 In the United States, the median life span
globin solubility test result. On alkaline electrophoresis, Hb D for men is 60 years and for women, 68 years.27
and Hb G have the same mobility as Hb S (see Figure 26-7).
Hb D and Hb G can be separated from Hb S on citrate agar at Laboratory Diagnosis
pH 6.0 (see Figure 26-7). These variants should be suspected The complete blood count (CBC) shows a mild normocytic,
whenever a hemoglobin is encountered that migrates in the S normochromic anemia with many of the features associated
position on alkaline electrophoresis and has a negative result with sickle cell anemia. The hemoglobin level is usually 11 to
on the hemoglobin solubility test. In the homozygous state 13 g/dL, and the reticulocyte count is 3% to 5%. On the periph-
(Hb DD), there is greater than 95% Hb D with normal amounts eral blood film, there are a few sickle cells, target cells, and
of Hb A2 and Hb F.27 Hb DD can be confused with the com- intraerythrocytic crystalline structures. Crystalline aggregates of
pound heterozygous state for Hb D and 0-thalassemia. The hemoglobin (SC crystals) form in some cells, where they pro-
two disorders can be differentiated on the basis of RBC indices, trude from the membrane (Figure 26-10).77,80 Hb SC crystals
levels of Hb A2, and family studies. often appear as a hybrid of Hb S and Hb C crystals. They are
Hb D and Hb G are asymptomatic in the heterozygous state. longer than Hb C crystals but shorter and thicker than Hb S
Hb D disease (Hb DD) is marked by mild hemolytic anemia polymers and are often branched.
and chronic nonprogressive splenomegaly. No treatment is The result of the hemoglobin solubility screening test is
required. positive because of the presence of Hb S. Electrophoretically,
Hb C and Hb S migrate in almost equal amounts (45%) on
cellulose acetate, and Hb F is normal. Hb C is confirmed on
COMPOUND HETEROZYGOSITY WITH
citrate agar at an acid pH, where it is separated from Hb E and
HEMOGLOBIN S AND ANOTHER
Hb O. Hb A2 migrates with Hb C, and its quantitation is of no
-HEMOGLOBIN MUTATION
consequence in Hb SC disease. Determination of Hb A2
Compound heterozygosity is the inheritance of two different becomes vital, however, if a patient is suspected of having Hb
mutant genes that share a common genetic locus, in this case C concurrent with -thalassemia (see Chapter 27).
the -globin gene locus.77-80 Because there are two -globin
genes, these compound heterozygotes have inherited Hb S Treatment and Prognosis
from one parent and another chain hemoglobinopathy or Therapy similar to that for SCD is given to individuals with Hb
thalassemia from the other parent. Compound heterozygosity SC disease.73
of Hb S with Hb C, Hb D, Hb O, or -thalassemia may produce
hemolytic anemia of variable severity. Inheritance of Hb S with Hemoglobin S-Thalassemia
other hemoglobins, such as Hb E, Hb G-Philadelphia, and Hb Compound heterozygosity for Hb S and -thalassemia is the
Korle Bu, causes disorders of no clinical consequence.77 most common cause of sickle cell syndrome in patients of
Mediterranean descent and is second to Hb SC disease among
Hemoglobin SC all compound heterozygous sickle disorders. Hb S-thal
Hb SC is the most common compound heterozygous syn- usually causes a clinical syndrome resembling that of mild or
drome that results in a structural defect in the hemoglobin moderate sickle cell anemia. The severity of this compound
molecule in which different amino acid substitutions are found heterozygous condition depends on the chain production of
on each of two -globin chains. At the sixth position, glutamic the affected -thalassemia gene. If there is no -globin chain
acid is replaced by valine (Hb S) on one -globin chain and production from the -thalassemia gene (S0-thal), the clini-
by lysine (Hb C) on the other -globin chain. The frequency cal course is similar to that of homozygous sickle cell anemia.
of Hb SC is 25% in West Africa. The incidence in the United If there is production of -globin (S+-thal), patients tend to
States is approximately 1 in 833 births.77,81 have a milder condition than patients with Hb SC. These
patients can be distinguished from individuals with sickle cell
Clinical Features trait because of the presence of greater amounts of Hb S than
Hb SC disease resembles a mild SCD. Growth and develop- of Hb A, increased levels of Hb A2 and Hb F, microcytosis from
ment are delayed compared with normal children. Unlike Hb the thalassemia, hemolytic anemia, abnormal peripheral blood
SS, Hb SC usually does not produce significant symptoms until morphology, and splenomegaly (see Chapter 27).17,77
382 PART IV Hematopathology: Erythrocyte Disorders

A B
Figure 26-10 A, Peripheral blood film for a patient with hemoglobin (Hb) SC (1000). Note intraerythrocytic, blunt-ended SC crystals and target cells.
B, Peripheral blood film for a patient with Hb SC showing two cells containing SC crystals and hypochromic, target, and folded cells (1000). (Courtesy Ann
Bell, University of Tennessee, Memphis.)

Hemoglobin SD and Hemoglobin Hemoglobin C-Harlem


SG-Philadelphia Hb C-Harlem has two substitutions on the chain: the sickle
Hb SD is a compound heterozygous and Hb SG-Philadelphia mutation and the Korle Bu mutation. Patients heterozygous for
a double heterozygous sickle cell syndrome.17,84 Hb SG- only Hb C-Harlem are asymptomatic. The compound hetero-
Philadelphia is asymptomatic, because Hb G is associated zygous Hb SHb C-Harlem state resembles Hb SS clinically.
with an gene mutation that still allows for sufficient Hb A to Hb C-Harlem yields a positive result on the hemoglobin solu-
be produced. Hb SD syndrome may cause a mild to severe bility test and migrates to the Hb C position on cellulose
hemolytic anemia because both chains are affected. Some acetate electrophoresis at an alkaline pH and to the Hb S
patients with Hb SD may have severe vasoocclusive complica- position on citrate agar electrophoresis at an acid pH.
tions. The Hb D syndrome in African Americans is usually Table 26-2 summarizes common clinically significant
due to the interaction of Hb S with Hb D-Los Angeles hemoglobinopathies, including general characteristics and
(Hb D-Punjab). treatment options.
The peripheral blood film findings for Hb SD disease are
comparable to those seen in less severe forms of Hb SS disease.
HEMOGLOBIN M
Because Hb D and Hb G co-migrate with Hb S on cellulose
acetate electrophoresis at an alkaline pH, citrate agar electro- Hb M is caused by a variety of mutations in the , , and
phoresis at an acid pH is necessary to separate Hb S from Hb genes, all of which result in the production of methemoglobin
D and Hb G. The clinical picture is valuable in differentiating hence the Hb M designation.84,85 These genetic mutations result
Hb SD and Hb SG. The treatment for Hb SD disease is similar in a structural abnormality in the globin portion of the mol-
to that for patients with SCD and is administered according to ecule. Most M hemoglobins involve a substitution of a tyrosine
the severity of the clinical condition. amino acid for either the proximal (F8) or the distal (E7) histi-
dine amino acid in the , , or chains. These substitutions
Hemoglobin SO-Arab cause heme iron to autooxidize, which results in methemoglo-
Hb SO-Arab is a rare compound heterozygous hemoglo binemia. Hb M has iron in the ferric state (Fe3+) and is unable
binopathy that causes severe chronic hemolytic anemia with to carry oxygen, which produces cyanosis. At least five hemo-
vasoocclusive episodes.5,17,84 Hb SO-Arab can be mistaken globin variants affecting the or chains have been classified
for Hb SC on cellulose acetate electrophoresis at an alkaline as M hemoglobins: Hb M-Boston, Hb M-Iwate, Hb M-Saskatoon,
pH because Hb C and Hb O-Arab migrate together; however, Hb M-Milwaukee-1, and Hb M-Milwaukee-2 (Hyde Park), all
differentiation is easily made on citrate agar at an acid pH. named for the locations in which they were discovered. Two
Therapy for these patients is similar to that for patients variants affect the chain, Hb FM-Osaka and Hb FM-Fort
with SCD. Ripley, but symptoms disappear when Hb A replaces Hb F at
3 to 6 months of age.
Hemoglobin S-Korle Bu Hb M variants have altered oxygen affinity and are inherited
Hb Korle Bu is a rare hemoglobin variant.17 When inherited as autosomal dominant disorders. When Hb M is exposed to
with Hb S, it interferes with lateral contact between Hb S fibers certain oxidants, such as sulfonamides or phenacetin, a choco-
by disrupting the hydrophobic pocket for 6 valine, which late brown color develops. Affected individuals have 30% to
inhibits Hb S polymerization. The compound heterozygous 50% methemoglobin (healthy individuals have less than 1%)
condition Hb S-Korle Bu is asymptomatic. and may appear cyanotic. Methemoglobin causes the blood

TABLE 26-2 Common Clinically Significant Hemoglobinopathies


Hemoglobin
Hemoglobin Abnormal Structural Groups Primarily Solubility Red Blood Cell Symptoms/Organ
Disorder Hemoglobin Defect Affected Test Results Hemoglobins Present Morphology Defects Treatment
6GluVal
Sickle cell anemia Hb S 22 African, African Positive 0% Hb A, >80% Hb S, Sickle cells, Vasoocclusion, Transfusions,
(homozygous) American, Middle 1-20% Hb F, 2-5% target cells, bacterial infections, antibiotics,
Eastern, Indian, Hb A2 nucleated RBCs, hemolytic anemia, analgesics,
Mediterranean polychromasia, aplastic episodes; bone marrow
Howell-Jolly bodies, bones, lungs, liver, transplants,
basophilic stippling, spleen, penis, eyes, hydroxyurea
spherocytes central nervous
system, urinary tract
Hb C disease Hb C 226GluLys African, African Negative 0% Hb A, >90% Hb C, Hb C crystals, target Mild splenomegaly, Usually none,
(homozygous) American <7% Hb F, 2% Hb A2 cells, nucleated mild hemolysis antibiotics
RBCs, occasionally
some microcytes
Hb C-Harlem (Hb Hb C-Harlem 226GluVal and Rare, so uncertain; Positive Migrates with Hb C at Target cells Compound Usually none
C-Georgetown); 2273AspAsn African, African alkaline pH; migrates heterozygotes with
occurs as compound on same gene American with Hb S at acid pH Hb S have
heterozygosity with symptoms similar to
Hb S (Hb S/Hb Hb SS
C-Harlem)
Hb E disease Hb E 2226GluLys Southeast Asian, Negative 0% Hb A, 95% Hb E, Target cells, Mild anemia, mild Usually none
(homozygous) African, African 2-4% Hb A2; migrates microcytes splenomegaly, no
American with Hb A2, Hb C, and symptoms
Hb O at alkaline pH
Hb O-Arab Hb O 22121GluLys Kenyan, Israeli, Negative 0% Hb A, 95% Hb O, Target cells Mild splenomegaly Usually none
(homozygous) Egyptian, 2-4% Hb A2; migrates
Bulgarian, African with Hb A2, Hb C, and
American Hb E at alkaline pH
Hb D disease (rare Hb D-Los 22121GluGln Middle Eastern, Negative 95% Hb D, normal Hb A2 Target cells Mild hemolytic anemia, Usually none
homozygous) Angeles, Hb Indian and Hb F; migrates with mild splenomegaly
D-Punjab Hb S at alkaline pH
Hb G disease (rare Hb G, Hb 268AsnLys2 African American, Negative 95% Hb G, normal Hb A2 Target cells Mild hemolytic anemia, Usually none
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin)

homozygous) G-Philadelphia Ghanaian and Hb F; migrates with mild splenomegaly


S at alkaline pH
383
384

TABLE 26-2 Common Clinically Significant Hemoglobinopathiescontd


Hemoglobin
Hemoglobin Abnormal Structural Groups Primarily Solubility Red Blood Cell Symptoms/Organ
Disorder Hemoglobin Defect Affected Test Results Hemoglobins Present Morphology Defects Treatment
Hb SC disease Hb S, Hb C 226GluVal and Same as Hb S Positive 45% Hb S, 45% Hb C, Sickle cells, Hb SC Same as those for Hb Similar to that
226GluLys; 2-4% Hb A2, 1% Hb F crystals, target SS except milder for Hb S
separate genes cells
Hb S-Thalassemia Hb S + 226GluVal and Same as Hb S Positive Hb S variable, some Hb A Sickle cells, target Hemolytic anemia, Similar to that
-thalassemia 0 or + in +, increased Hb A2 cells, microcytes splenomegaly for Hb S;
mutation and Hb F varies
PART IV Hematopathology: Erythrocyte Disorders

depending on
amount of Hb
A present
Hb SD disease Hb S, Hb D 226GluVal and Same as Hb S Positive 45% Hb S, 45% Hb D, Sickle cells, target Similar to those for Hb Similar to that
22121GluGln 2-4% Hb A2, 1% Hb F; cells S but milder for Hb S but
Hb S and D co-migrate less intensive
at alkaline pH
Hb SG Hb S, Hb G 226GluVal and Same as Hb S Positive 45% Hb S, 45% Hb G, Target cells No symptoms Usually none
268AsnLys2 2-4% Hb A2, 1% Hb F;
Hb S and G co-migrate
at alkaline pH
Hb SO Hb S, Hb O 226GluVal and Same as Hb S Positive 45% Hb S, 45% Hb O, Sickle cells, target Similar to those for Hb Similar to that
22121GluLys 2-4% Hb A2, 1% Hb F cells S for Hb S

Asn, Asparagine; Asp, aspartic acid; Gln, glutamine; Glu, glutamic acid; Hb, hemoglobin; Lys, lysine; Val, valine.
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 385

sample to appear brown. Heinz bodies may be seen sometimes such as drug ingestion and exposure to heat or cold. The hemo-
on wet preparations, because methemoglobin causes globin globin precipitates in the RBC as Heinz bodies. The precipi-
chains to precipitate (see Figure 14-11). Diagnosis is made by tated hemoglobin attaches to the cell membrane, causing
spectral absorption of the hemolysate or by hemoglobin elec- clustering of band 3, attachment of autologous immunoglobu-
trophoresis. The absorption spectrum peaks are determined at lin, and macrophage activation. In addition, Heinz bodies
various wavelengths. The unique absorption range of each Hb can be trapped mechanically in the splenic sieve, which
M variant is identified when these are compared with the spec- shortens RBC survival. The oxygen affinity of these cells is also
trum of normal blood. abnormal.
Before electrophoresis, all hemoglobin types are converted The most prevalent unstable hemoglobin is Hb Kln. Other
to methemoglobin by adding potassium cyanide to the sample unstable hemoglobins include Hb Hammersmith, Hb Zurich,
so that any migration differences observed are only due to an Hb Gunn Hill, and Hb Cranston. Because of the large vari-
amino acid substitution, not differences in iron states. On cel- ability in the degree of instability in these hemoglobins, the
lulose acetate, Hb M migrates slightly more slowly than Hb A. extent of hemolysis varies greatly. For some of the variants,
The electrophoresis should be performed on agar gel at pH 7.1 such as Hb Zurich, the presence of an oxidant is required for
for clear separation. Further confirmation may be obtained any significant hemolysis to occur.
using HPLC or deoxyribonucleic acid (DNA)-based globin
gene analysis. No treatment is necessary. Diagnosis is essential Laboratory Diagnosis
to prevent inappropriate treatment for other conditions, such The RBC morphology varies. It may be normal or show slight
as cyanotic heart disease. hypochromia and prominent basophilic stippling, which
possibly is caused by excessive clumping of ribosomes. Before
splenectomy, the hemoglobin level ranges from 7 to 12 g/dL
UNSTABLE HEMOGLOBIN VARIANTS
with a 4% to 20% reticulocyte count. After splenectomy,
Unstable hemoglobin variants result from genetic mutations anemia is corrected, but reticulocytosis persists. Heinz bodies
to globin genes creating hemoglobin products that precipitate can be shown using a supravital stain (see Figure 14-11). After
in vivo, producing Heinz bodies and causing a hemolytic splenectomy, Heinz bodies are larger and more numerous.
anemia.84,85 More than 200 variants of unstable hemoglobin Many patients excrete dark urine that contains dipyrrole.
exist. The majority of these are chain variants; most others Many unstable hemoglobins migrate in the normal AA
are chain variants. Only a few are and chain variants. pattern and thus are not detected on electrophoresis. Other
Most unstable hemoglobin variants have no clinical signifi- tests used to detect unstable hemoglobins include the isopro-
cance, although the majority have an increased oxygen affinity. panol precipitation test, which is based on the principle that
About 25% of unstable hemoglobins are responsible for hemo- an isopropanol solution at 37 C weakens the bonding forces
lytic anemia, which varies from compensated mild anemia to of the hemoglobin molecule. If unstable hemoglobins are
severe hemolytic episodes. present, rapid precipitation occurs in 5 minutes, and heavy
At one time, the anemia was referred to as congenital nons- flocculation occurs after 20 minutes. Normal hemoglobin does
pherocytic hemolytic anemia or congenital Heinz body anemia. This not begin to precipitate until approximately 40 minutes. The
disorder is more properly called unstable hemoglobin disease. The heat denaturation test also can be used. When incubated at
syndrome appears at or just after birth depending on the globin 50 C for 1 hour, heat-sensitive unstable hemoglobins show a
chains involved. It is inherited in an autosomal dominant flocculent precipitation, whereas normal blood shows little or
pattern. All patients are heterozygous; apparently the homozy- no precipitation. Significant numbers of Heinz bodies appear
gous condition is incompatible with life. The instability of the after splenectomy, but even in individuals with intact spleens,
hemoglobin molecule has several causes: (1) substitution of a with longer incubation and the addition of an oxidative sub-
charged for an uncharged amino acid in the interior of the stance such as acetylphenylhydrazine, unstable hemoglobins
molecule, (2) substitution of a polar for a nonpolar amino acid form more Heinz bodies than does the blood from individuals
in the hydrophobic heme pocket, (3) substitution of an amino with normal hemoglobins. Newer techniques, such as isoelec-
acid in the and chains at the intersubunit contact points, tric focusing, can resolve many hemoglobin mutations with
(4) replacement of an amino acid with proline in the helix only a slight alteration in their isoelectric point, and globin
section of a chain, and (5) deletion or elongation of the chain analysis can be performed by HPLC or DNA-based
primary structure. globin gene analysis.

Clinical Features Treatment and Prognosis


The unstable hemoglobin disorder is usually detected in early Patients are treated to prevent hemolytic crises. In severe cases,
childhood in patients with hemolytic anemia accompanied by the spleen must be removed to reduce sequestration and rate
jaundice and splenomegaly. Fever or ingestion of an oxidant of removal of RBCs. Because unstable hemoglobin disease is
exacerbates the hemolysis. The severity of the anemia depends rare, prognosis in the affected individuals is unclear. Patients
on the degree of instability of the hemoglobin molecule. The are cautioned against the use of sulfonamides and other
unstable hemoglobin precipitates in vivo and in vitro in oxidant drugs. They also should be informed of the potential
response to factors that do not affect normal hemoglobins, for febrile illnesses to trigger a hemolytic episode.
386 PART IV Hematopathology: Erythrocyte Disorders

More than 115 variant hemoglobins with high oxygen affin-


HEMOGLOBINS WITH INCREASED AND
ity have been discovered. Such hemoglobins fail to release
DECREASED OXYGEN AFFINITY
oxygen on demand, and hypoxia results. The kidneys sense the
More than 150 hemoglobin variants have been discovered to hypoxia and respond by increasing the release of erythro
have abnormal oxygen affinity.1,85-87 Most are high-affinity poietin, which leads to a compensatory erythrocytosis. These
variants and have been associated with familial erythrocytosis. variants differ from unstable hemoglobin, which also may
The remaining hemoglobin variants are characterized by low have abnormal oxygen affinity, in that they do not precipitate
oxygen affinity. Many of these are associated with mild to in vivo to produce hemolysis and there is no abnormal RBC
moderate anemia.2 morphology.
As described in Chapter 10, normal Hb A undergoes a Most individuals are asymptomatic and show no physical
series of allosteric conformational changes as it converts symptoms except a ruddy complexion. Erythrocytosis is usually
from a fully deoxygenated to a fully oxygenated form. These detected during routine examination, because the patient gen-
conformational changes affect hemoglobin function and its erally has a high RBC count, hemoglobin, and hematocrit. The
affinity for oxygen. When normal hemoglobin is fully deoxy- WBC count, platelet count, and peripheral blood film findings
genated (tense state), it has low affinity for oxygen and other are generally normal. In some cases, hemoglobin electropho-
heme ligands and high affinity for allosteric effectors, such resis may establish a diagnosis. An abnormal band that sepa-
as Bohr protons and 2,3-bisphosphoglycerate. In the oxygen- rates from the A band is present on cellulose acetate in some
ated (relaxed) state, hemoglobin has a high affinity for heme variants; however, if a band is not found, the diagnosis of
ligands, such as oxygen, and a low affinity for Bohr protons increased oxygen affinity cannot be ruled out. In some cases
and 2,3-bisphosphoglycerate. The transition from the tense to the abnormal hemoglobin can be separated by using citrate
the relaxed state involves a series of structural changes that agar (pH 6.0) or by gel electrophoresis. Measurement of oxygen
have a marked effect on hemoglobin function. If an amino acid affinity is required for definitive diagnosis.
substitution lowers the stability of the tense structure, the tran- Patients with high-oxygen-affinity hemoglobins live normal
sition to the relaxed state occurs at an earlier stage in ligand lives and require no treatment. Diagnosis should be made to
binding, and the hemoglobin has increased oxygen affinity and avoid unnecessary treatment of the erythrocytosis as a myelo-
decreased heme-heme interaction or cooperativity (see Chapter proliferative neoplasm or a secondary erythrocytosis.
10). One example of a chain variant is Hb Kempsey. This
unstable hemoglobin variant has amino acid substitutions at
sites crucial to hemoglobin function. Hemoglobins with Decreased Oxygen Affinity
Hemoglobins with decreased oxygen affinity quickly release
Hemoglobins with Increased Oxygen Affinity oxygen to the tissues, which results in normal to decreased
The high-affinity variants, like other structurally abnormal hemoglobin concentration and slight anemia. The best known
hemoglobins, show an autosomal dominant pattern of inheri- of these hemoglobins is Hb Kansas, which has an amino acid
tance. Affected individuals have equal volumes of Hb A and substitution of asparagine by threonine at the 102nd position
the abnormal variant. Exceptions to this are compound hetero- of the chain. These hemoglobins may be present when cya-
zygotes for Hb Abruzzo and -thalassemia and for Hb Crete nosis and a normal arterial oxygen tension coexist, and most
and -thalassemia, in which the proportion of abnormal may be detected by starch gel electrophoresis, HPLC, or
hemoglobin is greater than 85%. DNA-based globin gene analysis.

SUMMARY
Hemoglobinopathies are genetic disorders of globin genes that The most clinically significant hemoglobinopathies are Hb SS, Hb
produce structurally abnormal hemoglobins with altered amino SC, and Hb S-thalassemia; Hb SS causes the most severe
acid sequences, which affect hemoglobin function and stability. disease.
Hb S is the most common hemoglobinopathy, resulting from a Individuals with sickle cell trait (Hb AS) are clinically
substitution of valine for glutamic acid at the sixth position of the asymptomatic.
globin chain, and primarily affects people of African descent. Sickle cell anemia (Hb SS) is a normocytic, normochromic anemia,
Hb S polymerizes in the RBC because of abnormal interaction with characterized by a single band in the S position on hemoglobin
adjacent tetramers when it is in the deoxygenated form, producing electrophoresis and a positive hemoglobin solubility test.
sickle-shaped RBCs. The median life expectancy of patients with SCD has been
In SCD (homozygous Hb SS), the polymerization of hemoglobin extended to approximately 50 years.
may result in severe episodic conditions; however, factors other Hb C and Hb E are the next most common hemoglobinopathies after
than hemoglobin polymerization may account for vasoocclusive Hb S and cause mild hemolysis in the homozygous state. In the
episodes in sickle cell patients. heterozygous states, these hemoglobinopathies are asymptomatic.
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 387

Hb C is found primarily in people of African descent. reticulocyte count, hemoglobin solubility testing, hemoglobin elec-
On peripheral blood films from patients with Hb CC, hexagonal trophoresis (acid and alkaline pH), and measurement of Hb A2 and
crystals may be seen with and without apparent RBC membrane Hb F levels.
surrounding them. Advanced techniques available for hemoglobin identification
Hb EE results in a microcytic anemia and is found primarily in include HPLC, capillary electrophoresis, and isoelectric focusing.
people of Southeast Asian descent.
Other variants, such as unstable hemoglobins and hemoglobins Now that you have completed this chapter, go back and
with altered oxygen affinity, can be identified, and many cause no read again the case study at the beginning and respond
clinical abnormality. to the questions presented.
Laboratory procedures employed to determine the presence of an
abnormal hemoglobin are CBC, peripheral blood film evaluation,

R E V I E W Q UESTIONS
1. A qualitative abnormality in hemoglobin may involve all a. Decreased production of chains
of the following except: b. Substitution of lysine for glutamic acid at the sixth
a. Replacement of one or more amino acids in a globin position of the chain
chain c. Substitution of tyrosine for the proximal histidine in
b. Addition of one or more amino acids in a globin chain the chain
c. Deletion of one or more amino acids in a globin chain d. Double amino acid substitution in the chain
d. Decreased production of a globin chain
7. A well-mixed specimen obtained for a CBC has a brown
2. The substitution of valine for glutamic acid at the sixth color. The patient is being treated with a sulfonamide for
position of the chain of hemoglobin results in hemoglo- a bladder infection. Which of the following could explain
bin that: the brown color?
a. Is unstable and precipitates as Heinz bodies a. The patient has Hb M.
b. Polymerizes to form tactoid crystals b. The patient is a compound heterozygote for Hb S and
c. Crystallizes in a hexagonal shape thalassemia.
d. Contains iron in the ferric (Fe3+) state c. The incorrect anticoagulant was used.
d. Levels of Hb F are high.
3. Patients with SCD usually do not exhibit symptoms until
6 months of age because: 8. Through routine screening, prospective parents discover
a. The mothers blood has a protective effect that they are both heterozygous for Hb S. What percentage
b. Hemoglobin levels are higher in infants at birth of their children potentially could have SCD?
c. Higher levels of Hb F are present a. 0%
d. The immune system is not fully developed b. 25%
c. 50%
4. Megaloblastic episodes in SCD can be prevented by pro- d. 100%
phylactic administration of:
a. Iron 9. Painful crises in patients with SCD occur as a result of:
b. Folic acid a. Splenic sequestration
c. Steroids b. Aplasia
d. Erythropoietin c. Vasoocclusion
d. Anemia
5. Which of the following is the most definitive test for Hb S?
a. Hemoglobin solubility test 10. The screening test for Hb S that uses a reducing agent, such
b. Hemoglobin electrophoresis at alkaline pH as sodium dithionite, is based on the fact that hemoglo-
c. Osmotic fragility test bins that sickle:
d. Hemoglobin electrophoresis at acid pH a. Are insoluble in reduced, deoxygenated form
b. Form methemoglobin more readily and cause a color
6. A patient presents with mild normochromic, normocytic change
anemia. On the peripheral blood film, there are a few c. Are unstable and precipitate as Heinz bodies
target cells, a rare nucleated RBC, and hexagonal crystals d. Oxidize quickly and cause turbidity
within and lying outside of the RBCs. Which abnormality
in the hemoglobin molecule is most likely?
388 PART IV Hematopathology: Erythrocyte Disorders

REFERENCES
1. Wang WC: Sickle cell anemia and other sickling syndromes. 21. de Jong K, Kuypers FA: Sulfhydral modifications alter scam-
In Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes blase activity in murine sickle cell disease. Br J Haematol
clinical hematology, vol 1, ed 12, Philadelphia, 2009, 133:427-432, 2006.
Wolters Kluwer Health/Lippincott Williams & Wilkins, 22. Manodori AB, Barabino GA, Lubin BH, et al: Adherence
pp 1038-1082. of phosphatidylserine-exposing erythrocytes to endothelial
2. Patrinos GP, Giardine B, Riemer C, et al: Improvements matrix thrombomodulin. Blood 95:1293-1300, 2000.
in the HbVar database of human hemoglobin variants and 23. Stuart ML, Setty BN: Hemostatic alterations in sickle cell
thalassemia mutations for population and sequence varia- disease: relationships to disease pathophysiology. Pediatr
tion studies. Nucl Acids Res 32(database issue):D537-541, Pathol Mol Med 20:27-46, 2001.
2004. Available at: http://globin.cse.psu.edu/hbvar/menu. 24. Setty BN, Kulkarni S, Stuart MJ: Role of erythrocyte phospha-
html. Accessed May 3, 2010. tidylserine in sickle red cell-endothelial adhesion. Blood
3. Fishleder AJ, Hoffman GC: A practical approach to the detec- 99:1564-1571, 2002.
tion of hemoglobinopathies. Part II: the sickle cell disorders. 25. Kuypers FA, Styles LA: The role of secretory phospholipase A2
Lab Med 18:441-443, 1987. in acute chest syndrome. Cell Mol Biol 50:87-94, 2004.
4. Konotey-Ahulu FI: The sickle cell diseases. Clinical manifesta- 26. Neidlinger NA, Larkin SK, Bhagat A, et al: Hydrolysis of
tions including the sickle crisis. Arch Intern Med 133:611 phosphatidylserine-exposing red blood cells by secretory
619, 1974. phospholipase A2 generates lysophosphatidic acid and results
5. Beutler E: Disorders of hemoglobin structure: sickle cell in vascular dysfunction. J Biol Chem 281:775-781, 2006.
anemia and related abnormalities. In Lichtman MA, Beutler 27. Platt OS, Brambilla DJ, Rosse WF, et al: Mortality in sickle cell
E, Kipps TJ, et al, editors: Williams hematology, ed 7, New York, disease: life expectancy and risk factors for early death.
2006, McGraw-Hill, pp 667-700. N Engl J Med 330:1639-1644, 1994.
6. Serjeant GR: Historical review: the emerging understanding 28. Noguchi CT, Rodgers GP, Sergeant G, et al: Level of fetal
of sickle cell disease. Br J Haematol 112:3-18, 2001. hemoglobin necessary for treatment of sickle cell disease.
7. Park KW: Sickle cell disease and other hemoglobinopathies. N Engl J Med 318:96-99, 1988.
Int Anesth Clin 42:77-93, 2004. 29. Diggs LW: Sickle cell crises. J Clin Pathol 44:1-19, 1965.
8. Sickle Cell Disease Guideline Panel: Sickle cell disease: 30. Embury SH, Hebbel RP, Steinberg MH, et al. Pathogenesis of
screening, diagnosis, management, and counseling in new- vasoocclusion. In Embury SH, Hebbel RP, Mohandas N,
borns and infants, Clinical Practice Guideline, AHCPR Pub et al, editors: Sickle cell disease: basic principles and clinical
No 93-0562. Rockville, Md., 1993, Agency for Health Care practice, Philadelphia, 1994, Lippincott Williams & Wilkins,
Policy and Research. pp 311-323.
9. Weatherall DS: ABC of clinical haematology: the hereditary 31. Jandl JH: Blood: textbook of hematology, ed 2, Boston, 1996,
anaemias. BMJ 314:492-496, 1997. Little, Brown, pp 531-577.
10. Smith JA, Kinney TR, Sickle Cell Disease Guideline Panel: 32. Hebbel RP, Yamada O, Moldow CF, et al: Abnormal adher-
Sickle cell disease: guideline and overview. Am J Hematol ence of sickle erythrocytes to cultured vascular endothelium:
47:152-154, 1994. possible mechanism to microvascular occlusion in sickle cell
11. Shah A: Hemoglobinopathies and other congenital hemo- disease. J Clin Invest 65:154-160, 1980.
lytic anemia. Ind J Med Sci 58:490-493, 2004. 33. Bunn HF: Pathogenesis and treatment of sickle cell disease.
12. Wishner BC, Ward KB, Lattman EE, et al: Crystal structure of N Engl J Med 337:762-769, 1997.
sickle-cell deoxyhemoglobin at 5 A resolution. J Mol Biol 34. Hagar W, Vichinsky E: Advances in clinical research in sickle
98:179-194, 1975. cell disease. Br J Haematol 141:346-356, 2008.
13. Dykes G, Crepeau RH, Edeltein SJ: Three-dimensional recon- 35. Styles LA, Schalkwijk CG, Aarsman AJ, et al: Phospholipase
struction of the fibers of sickle cell haemoglobin. Nature A2 levels in acute chest syndrome of sickle cell disease. Blood
272:506-510, 1978. 87:2573-2578, 1996.
14. Dykes GW, Crepeau RH, Edelstein SJ: Three-dimensional 36. Styles LA, Aareman AJ, Vichinski EP, et al: Secretory phospho-
reconstruction of the 14-filament fiber of hemoglobin S. J Mol lipase A2 predicts impending acute chest syndrome in sickle
Biol 130:451-472, 1979. cell disease. Blood 96:3276-3278, 2000.
15. Crepeau RH, Edelstein SJ, Szalay M, et al: Sickle cell hemo- 37. Morris CR, Kuypers FA, Larkin S, et al: Patterns of arginine
globin fiber structure altered by alpha-chain mutation. Proc and nitrous oxide in patients with sickle cell disease with
Natl Acad Sci U S A 78:1406-1410, 1981. vaso-occlusive crisis and acute chest syndrome. J Pediatr Hae-
16. Phillips JA III, Kazazian HH Jr: Hemoglobinopathies and matol Oncol 22:515-520, 2000.
thalassemias. In Spivak JL, Eichner ER, editors: Fundamentals 38. Morris CR, Kato GJ, Poljakovic M, et al: Dysregulated arginine
of clinical hematology, ed 3, Baltimore: Johns Hopkins Univer- metabolism, hemolysis-associated pulmonary hypertension,
sity Press, 1993, pp 47-76. and mortality in sickle cell disease. JAMA 294:81-90, 2005.
17. Heeney M, Dover GJ: Sickle cell disease. In Orkin SH, Nathan 39. Hsu LL, Champion HC, Campbell-Lee SC, et al: Hemolysis
DG, Ginsberg D, et al, editors: Nathan and Oskis hematology in sickle cell mice causes pulmonary hypertension due to
of infancy and childhood, ed 7, Philadelphia, 2009, Saunders, global impairment in nitric oxide bioavailability. Blood
pp 949-1014. 109:3088-3098, 2007.
18. de Jong K, Larkin SK, Styles LA, et al: Characterization of the 40. Onyekwere OC, Campbell A, Teshome M, et al: Pulmonary
phosphatidleserine-exposing subpopulation of sickle cells. hypertension in children and adolescence with sickle cell disease.
Blood 98:860-867, 2001. Pediat Cardiol 29(2):309-312, 2008.
19. Yasin Z, Witting S, Palascak MB: Phosphatidylserine external- 41. Kelleher JF, Luban NLC, Cohen BJ, et al: Human serum
ization in sickle red blood cell: associations with cell age, parvovirus as a cause of aplastic crisis in sickle cell disease.
density, and hemoglobin F. Blood 102:365-370, 2003. Am J Dis Child 138:401-403, 1984.
20. de Jong K, Rettig MP, Low PS, et al: Protein kinase C activation 42. Wethers DL: Sickle cell disease in childhood: Part I. Labora-
induces phosphatidylserine exposure on red blood cells. Bio- tory diagnosis, pathophysiology and health maintenance. Am
chemistry 41:12562-12567, 2002. Family Physician 62:1013-1020, 2000.
CHAPTER 26 Hemoglobinopathies (Structural Defects in Hemoglobin) 389
43. Agular C, Vichinsky E, Neumayr L: Bone and joint disease in 63. Martin HS: Review: sickle cell disease: present and future
sickle cell disease. Hematol Oncol Clin North Am 19:929-941, treatment. Am J Med Sci 312:166-174, 1996.
viii, 2005. 64. Pinto FO, Roberts I: Cord blood stem cell transplantation for
44. Nagel RL: Innate resistance to malaria: the intraerythrocytic haemoglobinopathies. Br J Haematol 141:309-324, 2008.
cycle. Blood Cells 16:321-339, 1990. 65. Surbek D, Schoeberlein A, Wagner A: Perinatal stem cell and
45. Beutler E, Johnson C, Powars D, et al: Prevalence of glucose- gene therapy for hemoglobinopathies. Semin Fetal Neonatal
6-phosphate dehydrogenase deficiency in sickle cell disease. Med 13:282-290, 2008.
N Engl J Med 290:826-828, 1974. 66. Reid CD, Charache S, Lubin B, et al: Management and therapy
46. Steinberg MH, West MS, Gallagher D, et al: Effects of glucose-6 of sickle cell disease, ed 3, Pub No 95-2117. Bethesda, Md.,
phosphate dehydrogenase deficiency upon sickle cell anemia. 1995, National Heart, Lung and Blood Institute.
Blood 71:748-752, 1988. 67. Claster S, Vichinsky E: Managing sickle cell disease. BMJ
47. Welsh S: Hemoglobinopathies and thalassemias. The ABCs 327:1151-1155, 2003.
of Lab Evaluation. Clin Lab News 35(10), 2009. Available at: 68. Steinberg MH, Ballas SK, Brunson CY, et al: Sickle cell anemia
http://www.aacc.org/publications/cln/2009/october/Pages/ in septuagenarians. Blood 86:3997-3998, 1995.
Series1009.aspx#. Accessed May 5, 2010. 69. Michlitsch J, Azimi M, Hoppe C, et al: Newborn screening
48. Clarke GM, Higgins TN: Laboratory investigation of hemo- for hemoglobinopathies in California. Pediatr Blood Cancer
globinopathies and thalassemias: review and update. Clin 52:486-490, 2009.
Chem 46:1284-1290, 2000. 70. Schlitt LE, Keital HG: Renal manifestations of sickle cell
49. Bargoma EM, Mitsuyashi JK, Larkin SK, et al: Serum C-reactive disease: a review. Am J Med Sci 239:773-778, 1960.
protein parallels secretory phospholipase A2 in sickle cell 71. Diggs LW, Bell A: Intraerythrocytic hemoglobin crystals in
disease patients with vaso-occlusive crisis or acute chest syn- sickle cell hemoglobin C disease. Blood 25:218-223, 1965.
drome. Blood 105:3384-3385, 2005. 72. Bell A: Homozygous C disease, Hematology Tech Sample H-71.
50. Walters PB, Fung EB, Killilea DW, et al: Oxidative stress and Chicago, 1974, American Society of Clinical Pathologists.
inflammation in iron-overloaded patients with beta- 73. Rabinovitch A: Hemoglobinopathy survey, set HG-B, Northfield,
thalassemia or sickle cell disease. Br J Haematol 135:254-263, Minn., 1992, CAP, pp 2-3.
2006. 74. Fairbanks VF, Gilchrist GS, Brimhall B, et al: Hemoglobin E
51. Gaston MH, Verter JI, Woods G, et al: Prophylaxis with oral trait reexamined: a cause of microcytosis and erythrocytosis.
penicillin in children with sickle cell disease. A randomized Blood 53:109-115, 1979.
trial. N Engl J Med 314:1593-1599, 1986. 75. Anderson HM, Ranney HM: Southeast Asian immigrants: the
52. Preboth M: Management of pain in sickle cell disease. Am new thalassemias in America. Semin Hematol 27:239-246,
Fam Physician 61:1-7, 2000. 1990.
53. Fung EB, Harmatz PR, Lee PD, et al: Increased prevalence 76. Brewer GJ, Iyengar V, Prasad AS: Clinical aspects of hemoglo-
of iron-overload associated endocrinopathy in thalassemia binopathies. In Bick RL, Bennett JM, Brynes RK, et al, editors:
verses sickle cell disease. Br J Haematol 135:574-582, 2006. Hematology: clinical and laboratory practice, St Louis, 1993,
54. Fung EB, Harmatz PR, Milet M, et al: Morbidity and mortality Mosby, pp 307-326.
in chronically transfused subjects with thalassemia and sickle 77. Randolph TR: Pathophysiology of compound heterozygotes
cell disease. Am J Hematol 82:255-265, 2007. involving hemoglobinopathies and thalassemias. Clin Lab Sci
55. Vichinsky E, Onyekwere O, Porter J, et al: A randomized 21(4):240-248, 2008.
comparison of deferasirox verses deferoxamine for the treat- 78. Hoffman GC: The sickling disorders. Lab Med 21:797-807,
ment of transfusional iron overload in sickle cell disease. 1990.
Br J Haematol 136:501-508, 2007. 79. Moewe C: Hemoglobin SC: a brief review. Clin Lab Sci 6:158-
56. Bunn HF, Forget BG: Sickle cell diseaseclinical and epide- 159, 1993.
miological aspects. In Bunn HF, Forget BG, editors: Hemoglo- 80. Nagel RL, Lawrence C: The distinct pathobiology of sickle
bin: molecular, genetic and clinical aspects, Philadelphia, 1986, cell-hemoglobin C disease: therapeutic implications. Hematol
Saunders, pp 503-510. Oncol Clin North Am 5:433-451, 1991.
57. Walters MC, Patience M, Leisenring M, et al: Barriers to bone 81. Lawrence C, Fabry ME, Nagel RL: The unique red cell hetero-
marrow transplant for sickle cell anemia. Biol Bone Marrow geneity of SC disease: crystal formation, dense reticulocytes
Transplant 2:100-104, 1996. and unusual morphology. Blood 78:2104-2112, 1991.
58. Bernaudin F, Socie G, Kuentz M, et al: Long-term results of 82. Platt OS, Thorington BD, Brambilla DJ, et al: Pain in sickle cell
related, myeloablative stem cell transplantation to cure sickle disease: rates and risk factors. N Engl J Med 325:11-16, 1991.
cell disease. Blood 110:2749-2756, 2007. 83. Steinberg MH: Review: sickle cell disease: present and future
59. Panepinto JA, Walters MC, Carreras J, et al: Matched-related treatment. Am J Med Sci 312:166-174, 1996.
donor transplantation for sickle cell disease: report from the 84. Safko R: Anemia of abnormal globin development
Center for International Blood and Transplant Research. Br J hemoglobinopathies. In Stiene-Martin EA, Lotspeich-
Haematol 137:479-485, 2007. Steininger C, Koepke JA, editors: Clinical hematology: principles,
60. Vermylen C, Cornu G, Ferster Y, et al: Haermatopoietic procedures, correlations, ed 2, Philadelphia, 1990, Lippincott
stem cell transplantation for sickle cell anaemia: the first 50 Raven Publishers, pp 192-216.
inpatients transplanted in Belgium. Bone Marrow Transplant 85. Steinberg M: Hemoglobins with altered oxygen affinity, unsta-
22:1-6, 1998. ble hemoglobins, M-hemoglobins, and dyshemoglobinemias.
61. Walters MC, Storb R, Patience M, et al: Impact of bone In Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes
marrow transplantation for symptomatic sickle cell disease: clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
an interim report. Multicenter investigation of bone marrow Health/Lippincott Williams & Wilkins, pp 1132-1142.
transplantation for sickle cell disease. Blood 95:1918-1924, 86. Williamson D: The unstable haemoglobins. Blood Rev 7:146-
2000. 163, 1993.
62. Walters MC, Patience M, Leisenring M, et al: Collaborative 87. Bunn HF: Hemoglobinopathy due to abnormal oxygen
multicenter investigation of marrow transplantation for binding. In Bunn HF, Forget BG, editors: Hemoglobin: molecu-
sickle cell disease: current results and future directions. Biol lar, genetic and clinical aspects, Philadelphia, 1986, Saunders,
Bone Marrow Transplant 3:310-315, 1997. pp 595-616.
27 Thalassemias
Rakesh P. Mehta and Elaine M. Keohane

OUTLINE OBJECTIVES
Definitions and History After completion of this chapter, the reader will be able to:
Epidemiology
Genetic Control of Globin 1. Describe the hemoglobin (Hb) defect found in 8. Describe the treatment of homozygous -
Synthesis thalassemias. thalassemias, the risks involved, and the reason it is
Categories of 2. Name the chromosomes that contain the -globin necessary to monitor iron levels.
Thalassemia gene and the -globin gene clusters and the globin 9. Correlate the clinical syndromes of -thalassemia
Pathophysiology chains produced by each. with the number of genes present.
Genetic Defects Causing 3. Discuss the geographic distribution of thalassemia 10. Recognize the laboratory findings associated with
Thalassemia and explain its association with malaria. various -thalassemia syndromes.
-Globin Gene Cluster 4. Explain the pathophysiologic effects caused by 11. Justify genetic counseling for those of Southeast
Thalassemias the imbalance of globin chain synthesis in Asian descent who have the 0-thalassemia
Clinical Syndromes of
thalassemia. haplotype.
-Thalassemia
5. List the clinically defined thalassemic syndromes 12. Describe the clinical syndromes of thalassemia
Other Thalassemias
Caused by Defects in associated with genetic defects of the -globin gene associated with structural hemoglobin variants.
the -Globin Gene cluster and describe the clinical expression of each 13. Specify the tests that may be helpful in screening for
Cluster heterozygous and homozygous form. -thalassemia minor.
Screening for 6. Describe the type of genetic mutations that result in 14. Correlate the results of hemoglobin electrophoresis
-Thalassemia Minor -thalassemias, including hereditary persistence of or high-performance liquid chromatography and
-Thalassemias fetal hemoglobin (HPFH). quantitation of Hb F and Hb A2 with the various
Clinical Syndromes of 7. Recognize the pattern of laboratory findings in thalassemia syndromes.
-Thalassemia heterozygous and homozygous -thalassemias, 15. Differentiate -thalassemia minor from iron defi-
Thalassemia Associated including HPFH. ciency anemia.
with Structural
Hemoglobin Variants
Hemoglobin
SThalassemia
Hemoglobin
CThalassemia CASE STUDY
Hemoglobin
EThalassemia After studying the material in this chapter, the reader should be able to respond to the following
Diagnosis of Thalassemia case study:
History and Physical
A 24-year-old male medical student in the United States was found to have a hemoglobin of
Examination
Laboratory Findings 10.2g/dL in a hematology laboratory class. During discussion of the family history with this
Differential Diagnosis of student, a hematologist at the university discovered that his mother had always been anemic, had
Thalassemia Minor and periodically been given iron therapy, and had a history of several acute episodes of gallbladder
Iron Deficiency Anemia disease (attacks). Both of the students parents had been born in Sicily. A cousin on his mothers
side had two children who died of thalassemia major at the ages of 4 and 5 years, and had a third
young daughter with thalassemia major who was being treated with regular blood transfusions.
The students laboratory test results were as follows:
Continued

The foundation for this chapter is the work of Martha Payne. The authors would like to express their gratitude for the continued opportunity to
amend her fine endeavor.

390
CHAPTER 27 Thalassemias 391

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

Patient Results Reference Range


12
RBCs ( 10 /L) 5.74 4.60-6.00
Hb (g/dL) 10.2 14.0-18.0
Hct (%) 35 40-54
MCV (fL) 61.0 80-100
MCH (pg) 17.8 26-32
MCHC (g/dL) 29.1 32-36

Peripheral blood RBCs exhibited moderate microcytosis, 2. What laboratory values helped confirm the diagnosis?
slight hypochromia, and slight poikilocytosis with occasional 3. From what other disorders should this anemia be differ-
target cells, and several RBCs had basophilic stippling. Hb A2 entiated? What laboratory tests would be helpful? Why is
was 4.9% by high-performance liquid chromatography differentiation important?
(HPLC) (reference range, 0% to 3.5%). Serum ferritin level 4. If this individual was planning to have children, what
was 320ng/mL (reference range, 15 to 400ng/mL). genetic counseling should be done?
1. Why was the family history so important in this case, and
what diagnosis did it suggest?

chain of Hb A (22) is impaired. Thalassemias are named


DEFINITIONS AND HISTORY
according to the chain with reduced or absent synthesis.
The thalassemias are a diverse group of inherited disorders
caused by genetic alterations that reduce or preclude the syn-
EPIDEMIOLOGY
thesis of one or more of the globin chains of the hemoglobin
(Hb) tetramer. Since thalassemia was first described in 1925 Although approximately 7% of the worlds population has a
by Cooley and Lee,1 investigators have worked to define and thalassemia or hemoglobinopathy, the distribution of thalas-
better understand this fascinating but often confusing condi- semia is concentrated in the thalassemia belt that extends
tion. Cooley and Lee1 described four children with anemia, from the Mediterranean east through the Middle East and India
splenomegaly, mild hepatomegaly, and mongoloid facies. to Southeast Asia and south to Northern Africa (see Figure
These characteristics would later become typical findings in 26-3).4,5 The carrier frequency of -thalassemia depends on the
young children with untreated -thalassemia major, often region, with Sardinia, Cyprus, and Greece having the highest
referred to as Cooleys anemia. Seven years later, Whipple and frequency in Europe (6% to 19%) and India, Thailand, and
Bradford published a paper outlining the detailed autopsy Indonesia having the highest frequency in Asia and Southeast
studies of children who died of this disorder.2 Because of the Asia (0.3% to 15%).5 The carrier frequency of -thalassemia
high incidence of patients of Mediterranean descent with this varies considerably. In Europe, Cyprus has the highest carrier
disorder, Whipple called the disease Thalassic (Greek for great frequency at 14%. The carrier frequency reaches 50% to 60%
sea) anemia, which was subsequently changed to thalassemia.2 in Eastern Saudi Arabia and parts of Asia and Africa, and may
Several investigators in the 1940s demonstrated the genetic be as high as 75% to 80% in certain groups in Nepal, India,
basis for this anemia and were able to show that in patients Thailand, and Papua New Guinea.5
who were homozygous for this condition (thalassemia major) The geographic location of the thalassemia belt coincides
the disease had a severe course. The heterozygotes, however, with areas in which malaria is prevalent (see Figure 26-3).
not only were carriers, but also had a milder anemia (thalas- Thalassemia minor (heterozygous thalassemia) appears to
semia minor). In the 1950s, thalassemias resulting from defects impart resistance to malaria. This allowed the selective advan-
in the chain were described.2 Together the thalassemias are tage that established thalassemia in high frequency in areas in
the most common single-gene disorder in humans. The clinical which malaria is endemic.6,7 Several case-control studies evalu-
outcomes associated with the many genetic mutations are ated the incidence of thalassemia in subjects with severe
extremely wide ranging, with certain defects causing no anemia malaria compared with a control population and consistently
and other defects leading to fetal death. Thalassemia results found a lower incidence of thalassemia in the population with
from a reduction in the rate of synthesis of one or more of malaria than in the population without malaria. One study
the globin chains. This decreased globin production leads to demonstrated that the risk of death from malaria was 40%
imbalanced globin chain synthesis, defective hemoglobin pro- lower in patients with / thalassemia and more than 60%
duction, and damage to the red blood cells (RBCs) or their lower in those with / thalassemia.7 The mechanism of
precursors by the excess accumulation of the globin chain that this resistance is still not fully elucidated; however, two major
is not affected.3 Usually, the synthesis of either the or the theories have been put forward: defective growth of the parasite
392 PART IV Hematopathology: Erythrocyte Disorders

Figure 27-1 Chromosome organization of globin genes and their expression during development. The solid boxes indicate functional globin genes; the
open boxes indicate pseudogenes. The scale of the depicted chromosomal segments is in kilobases of DNA. The switch from embryonic to fetal hemoglobin
(Hb) occurs between 6 and 10 weeks of gestation, and the switch from fetal to adult hemoglobin occurs in the third trimester. (From Nathan DG, Orkin SH,
editors: Nathan and Oskis hematology of infancy and childhood, ed 5, Philadelphia, 1998, Saunders, pp 811-886.)

in the affected cell and increased phagocytosis of the infected TABLE 27-1 Reference Ranges for Normal Hemoglobins
cell.8,9 Although the exact mechanism is not known, the geo- (Hbs) in Adults
graphic distribution and the case-control studies corroborate
Hb A (22) 95-100%
the protective nature of the thalassemias in promoting resis-
Hb A2 (22) 0-3.5%
tance to malaria.
Hb F (22) 0-2%

GENETIC CONTROL OF GLOBIN SYNTHESIS


life, Hb A (22) is the predominant hemoglobin, with a low
The normal hemoglobin molecule is a tetramer (double dimer) percentage of Hb A2 (22) (Table 27-1; see Figure 10-6).3,11
of two -like chains ( or ) with two -like chains (, , , or The genes G and A code for two globin chains that differ
). Combinations of these chains produce six normal hemo- by a single amino acid (glutamic acid or alanine). Both of these
globins. Three are embryonic hemoglobins: Hb Gower-1 (22), globin chains are expressed in fetal hemoglobin, and it appears
Hb Gower-2 (22), and Hb Portland (22). The others are fetal that there is no functional difference between them. Similarly,
hemoglobin (Hb F, 22) and two adult hemoglobins (Hb A, the gene loci are duplicated on each chromosome 16 and are
22, and Hb A2, 22). The -like globin chain genes are denoted 1 and 2. Either of these genes can contribute to the
located on chromosome 16, whereas the -like globin gene two -globin chains in the hemoglobin tetramer, and no func-
cluster is on chromosome 11 (Figure 27-1). The -like globin tional difference seems to exist between the two. Interspersed
gene cluster contains the functional embryonic globin gene () between the functional genes on these chromosomes are four
and two functional adult genes (1 and 2). The -like globin functionless genelike loci or pseudogenes that are designated
gene cluster contains five functional genes, , G, A, , and .3,10 by the prefixed symbol . The purpose of these pseudogenes
These genes are positioned in the order that corresponds with is unknown.10 The organization of these genes on chromo-
their developmental stage of expression. During weeks 6 to 11 somes 16 and 11 is shown in Figure 27-1.
of gestation, the and genes are expressed, generating Hb An individual inherits one cluster of the five functional
Gower-1 (22). Expression of the genes begins close to the genes on chromosome 11 from each parent. The genotype for
eleventh week, and the gene is gradually switched off so that normal chain synthesis is designated /. Because the gene
Hb Gower-2 (22) is produced. Around the eleventh week, the loci are duplicated, two genes are inherited on each chromo-
gene is switched off, and chain synthesis begins. The some 16. A normal genotype is therefore designated /.
chains combine either with the remaining chains to make Hb
Portland (22) or with the chains to make Hb F (22), the
CATEGORIES OF THALASSEMIA
predominant hemoglobin of fetal life. Shortly before birth,
chain synthesis begins to decrease, whereas and chain syn- The thalassemias are divided into -thalassemias, which
thesis increases, so that by 6 months of age and through adult include all the disorders of reduced globin chains arising
CHAPTER 27 Thalassemias 393

from the -globin gene cluster on chromosome 11, and - TABLE 27-2 Genetic Designations in Thalassemia
thalassemias, which involve the 1 and 2 loci on chromosome
Designation Definition
16. Various defects of deletion (absence of a portion of a gene)
and nondeletion can cause each of these disorders. Even defects Designations for Normal -Globin and -Globin
that seem the same clinically are often heterogeneous at the Genes
genetic level.10,12 Normal gene; normal amount of chains produced;
The -thalassemias affect mainly the chain production, but one gene located on each chromosome 11
also may involve the , G, A, and chains. In the -thalassemias, Two normal genes on one chromosome (haplotype
0 is the designation for mutations in the gene in which no ); normal amount of chains produced; two
chains are produced. In the homozygous state (0/0), an genes located on each chromosome 16
individual does not produce Hb A (22). + is the designation
for mutations in the gene that result in a partial deficiency in Designations for the Major Thalassemic Genes
the production of the chains and a decrease in production of 0 gene mutation in which no chains are produced
Hb A. Some mutations in the gene lead to minimal reductions + gene mutation that results in 10-50% decrease in
in -globin production and are associated with mild or silent chain production
clinical states. The designation silent for silent carrier has been silent gene mutation that results in mildly decreased
used for those mutations. The designation 0 is used for muta- chain production
tions in the or genes in which no or chains are produced. 0 gene deletion or nondeletional mutation in which no
In the homozygous state (0/0), no Hb A (22) or Hb A2 or chains are produced; accompanied by some
(22) is produced. The designation Lepore indicates a fusion increase in chain production
of the and genes that produces Hb Lepore. Lepore gene fusion that produces a small amount of fusion
-Thalassemia, usually caused by gene deletion, involves product, hemoglobin Lepore; no or chains are
the gene complex (1 or 2 genes) on chromosome 16. The produced; accompanied by some increase in chain
designation + is used to indicate a deletion of either the 1 or production
the 2 gene in the gene complex (also called the haplo- HPFH Hereditary persistence of fetal hemoglobin; deletion
type), which results in a decreased production of chains from or nondeletional mutation in gene promoter in
that chromosome. The designation 0 is used for a deletion of which no or chains are produced; accompanied
both the 1 and 2 genes in the gene complex (also called by increase in chain production
the haplotype), which results in no production of chains 0 Deletion of both genes on one chromosome
from that chromosome.10,12 In cases in which there is a muta- (haplotype, ) that results in no chain production
tion in the gene rather than a deletion, -thalassemia can + Deletion of one gene on one chromosome (haplotype,
still result. The designation T is used for the conditions that ) that results in decreased chain production
result from these mutations.10 T Nondeletional mutation in one gene on one
The major gene designations in thalassemia are summa- chromosome (haplotype T) that results in
rized in Table 27-2. decreased chain production (T denotes
thalassemia)
PATHOPHYSIOLOGY
The clinical sequelae of thalassemia stem not only from a anemia is multifactorial and results from ineffective produc-
decreased production of a particular globin chain but also, tion and increased destruction. Typically, individuals with
more importantly, from an imbalance of all of globin synthe- -thalassemia are asymptomatic during fetal life and through
sis.10,13 For -thalassemia and -thalassemia, the resultant 4 to 6 months of age because Hb F (22) is the predominant
decrease in hemoglobin produces microcytic, hypochromic circulating hemoglobin. They become symptomatic only after
RBCs. The unequal production of either the - or -globin the to switch.11 In -thalassemia, accumulation of non-
chains leads to decreased RBC survival that is a significant chains has different consequences. Because chains are shared
contributor to the anemia. Importantly, the mechanism and by fetal and adult hemoglobins, decreased chain production
the degree of shortened RBC survival are different for the results in excess chains in the fetus and excess chains from
-thalassemias and -thalassemias. In the -thalassemias, the 6 months of age through adult life. In the fetus, the excess
unpaired, excess chains precipitate in the developing RBCs chains accumulate even if only one gene is deleted or non-
and damage their surfaces. These damaged RBCs are subse- functional. The amount of chain accumulation is propor-
quently destroyed by the macrophages in the bone marrow or tional to the number of deleted or nonfunctional genes.
in the circulation. The premature death of RBCs in the bone These excess chains form 4 tetramers of hemoglobin, called
marrow leads to ineffective erythropoiesis. In this situation, the Hb Bart. Because these tetramers are soluble, they do not pre-
bone marrow attempts to produce RBCs, but is not able to cipitate to any significant degree in the developing RBCs in the
release viable cells into the circulation. Some of these cells bone marrow and do not cause severe ineffective erythropoi-
leave the bone marrow only to be destroyed in the spleen esis. As the mature RBCs age in the circulation, however, these
through extravascular hemolysis. Therefore in -thalassemia the tetramers precipitate and form inclusion bodies. The spleen
394 PART IV Hematopathology: Erythrocyte Disorders

removes the cells with these inclusions, which results in mutations has been compiled for numerous ethnic groups, and
extravascular hemolysis. The RBCs are microcytic and hypo- this information is particularly relevant for prenatal diagnosis
chromic in -thalassemia owing to the decrease in hemoglobin of -thalassemia.5
synthesis.10,14 The anemia is a result not only of ineffective
production, but also of shortened RBC survival. Hb Bart (4) Clinical Syndromes of -Thalassemia
has a high affinity for oxygen and cannot adequately deliver Historically, -thalassemia has been divided into three clinical
oxygen to tissues. The fetus cannot survive with only Hb Bart syndromes: -thalassemia minor (heterozygous state), a mild
(found when all four genes are absent); hydrops fetalis devel- microcytic, hypochromic hemolytic anemia; -thalassemia
ops, and the fetus ultimately dies in utero or shortly after major (homozygous state), a severe anemia leading to depen-
birth.14 In the adult, chain tetramers form Hb H. This is dence on transfusions; and -thalassemia intermedia, with
discussed in the -thalassemia section later in the chapter. symptoms of severity between the other two.10,13,15 A fourth
syndrome (designated as silent carrier state) is now recognized
in patients with genetic changes in one of the two genes that
GENETIC DEFECTS CAUSING THALASSEMIA
result in no hematologic abnormalities (Table 27-3).15 The
Molecular cloning, sequencing of deoxyribonucleic acid clinical manifestations of the genetic changes depend on
(DNA), and functional analysis of cloned genes have revealed whether one or both of the genes are affected and the extent
many different types of defects at the molecular level.10,15 Types to which the gene or genes are expressed. Some mutations
of genetic defects that cause a decrease in or lack of production result in the absence of production of the chain, and these
of a particular globin chain and thalassemia include single mutations are designated as 0 genes. Other mutations lead to
nucleotide (or point) mutations, small insertions or deletions, production of the chains, but at a significantly reduced rate,
or large deletions. The mechanisms10 by which these mutations and these are designated as + genes.10 The range of chain
interfere with globin chain production include:
Reduced or absent transcription of messenger ribonucleic acid
(mRNA) due to mutations in the promoter region or initia-
TABLE 27-3 Some Clinical Syndromes of
tion codon of a globin gene
-Thalassemias
mRNA processing errors due to mutations that add or remove Genotype Hb A Hb A2 Hb F Hb Lepore
splice sites resulting in no globin chain or altered globin
Normal (Normal Hematologic Parameters)
chain production, as well as mutations in polyadenylation
/ N N N 0
sites that decrease mRNA stability
Translation errors due to mutations that change the codon
Silent Carrier State (Normal Hematologic
reading frame (frameshift mutations), substitute an incor-
Parameters)
rect amino acid codon (missense mutations), add a stop
silent/ N N N 0
codon causing premature chain termination (nonsense
mutations), or remove a stop codon, which results in an
Thalassemia Minor (Asymptomatic; Mild
elongated and unstable mRNA that produces a dysfunc-
Hemolytic Anemia; Microcytic, Hypochromic)
tional globin chain
+/ N to Sl 0
Deletion of one or more globin genes resulting in the lack of
0/ N to Sl 0
production of the corresponding globin chains10,15
0/ N to 5-20% 0
All of these heterogeneous genetic defects or mutations
Lepore/ 5-15%
cause a decrease in or lack of synthesis of a globin chain, result-
ing in the thalassemia syndromes.
Thalassemia Major (Severe Transfusion-
Dependent Hemolytic Anemia; Microcytic,
-GLOBIN GENE CLUSTER THALASSEMIAS Hypochromic)
+/+ V 0
Modern advances in genetic technology have revealed the het-
+/0 V 0
erogeneity of the -globin gene cluster DNA defects that lead
0/0 0 V 0
to the clinical syndrome of -thalassemia.15 More than 200
Lepore/Lepore 0 0 75% 25%
genetic abnormalities have been discovered in the -globin
gene cluster, including mutations in each of the , , and
genes or in combined sections of the -globin gene cluster.10,16
Thalassemia Intermedia (Moderate Hemolytic
Although numerous different mutations exist, a small subset
Anemia with Few Transfusion Requirements;
of mutations accounts for the majority of the mutant alleles
Microcytic, Hypochromic)
silent/silent 0
within a single ethnic group or geographic area in which -
0/0 0 0 100% 0
thalassemia is found.10 Because multiple mutations are present
0/0 0 N 0
in each population, most individuals with severe -thalassemia
are genetic compound heterozygotes for two different thalas- , Increased; , decreased; 0, absent; Hb, hemoglobin; N, normal; Sl, slight;
semia mutations. The frequency of specific -thalassemia V, variable.
CHAPTER 27 Thalassemias 395
production in these + gene mutations varies from 10% to 50% can vary from 3.5% to 8.0%. The Hb F level usually ranges
of normal chain synthesis. In addition, patients who are from 1% to 5%. Less common variants of -thalassemia minor
silent carriers have a normal hemoglobin level and mean cell exist, such as 0/ and Lepore/. Other rare variants have
volume (MCV), whereas other patients with defects that only atypical features, such as one variant (Dutch 0-thalassemia
minimally reduce -globin production have minimal changes minor) that shows the expected elevation in Hb A2 level but
in their RBC profiles. an Hb F level in the 5% to 20% range,19 and another variant
found in a Sardinian family in which the Hb A2 level is
Silent Carrier State of -Thalassemia normal.20
The designation silent includes the various heterogeneous
gene mutations that produce only a small decrease in produc- -Thalassemia Major
tion of the chains. The silent carrier state (silent/) results in -Thalassemia major is characterized by a severe anemia first
nearly normal - chain ratios and no hematologic abnormali- detected in early childhood as the to switch takes place.
ties.15,17 It was recognized through a study of families in which Normally at birth, the predominant hemoglobin is Hb F, but
the affected children had a more severe -thalassemia syn- over the next 6 months Hb A levels increase and Hb F levels
drome than a parent with typical -thalassemia minor.13,15,17 decrease (see Figure 10-6).10-12 Severe -thalassemia is usually
The parents had normal levels of Hb A2 and a slight micro diagnosed when the patient is between 6 months and 2 years
cytosis. Several patients who are homozygous for a silent of age, because the Hb A level does not increase as expected.
thalassemia gene (silent/silent) have been described. They have The natural history of this condition (in an untreated or inad-
a moderate microcytic, hypochromic anemia with a hemoglo- equately treated patient) is characterized by increasing hepato-
bin level in the range of 7 to 9g/dL. The Hb F values range splenomegaly, jaundice, and marked bone changes due to an
from 2% to 19%, and the Hb A2 level is elevated to the range expanded marrow cavity caused by extreme erythroid hyper-
normally seen in individuals with thalassemia minor.18 plasia. Skull radiographs may demonstrate a typical hair on
end appearance. Radiographs of the long bones may exhibit
-Thalassemia Minor (-Thalassemia Trait) a lacy appearance from marrow expansion, which causes a thin
-Thalassemia minor (also called -thalassemia trait) results bony cortex that predisposes to pathologic fracture (Figure
when one of the two genes that produce the -globin chains is 27-3). A typical facies results, with prominence of the forehead
defective (heterozygous state). It usually presents as a mild, (also known as frontal bossing), cheekbones, and upper jaw.
asymptomatic anemia.10,12,15 One gene is affected by a muta- Physical growth and development are delayed.10,12
tion that decreases or abolishes its function, whereas the other In untreated -thalassemia major, the hemoglobin level can
gene is normal. The hemoglobin ranges from 10 to 13g/dL, fall to 2 or 3g/dL.10 On evaluation of the peripheral blood
and the RBC count is normal or slightly elevated. The RBCs are film, marked microcytosis and hypochromia are seen, with a
microcytic and hypochromic, with some degree of poikilocy- wide variety of RBC sizes (anisocytosis) and shapes (poikilo-
tosis, including target cells and elliptocytes. Basophilic stip- cytosis), including target cells, teardrop cells, and elliptocytes.
pling may be seen in the RBCs on a Wright-stained peripheral RBC fragments and microspherocytes are present as a direct
blood film (Figure 27-2). The bone marrow shows mild to consequence of unbalanced globin chain synthesis. Basophilic
moderate erythroid hyperplasia, with slight ineffective erythro- stippling and many nucleated RBCs are also found (Figure
poiesis. Hepatomegaly and splenomegaly are seen in a few 27-4). The MCV is very low. The reticulocyte count ranges from
patients. In the most common -thalassemia minor syndromes 2% to 8%, which is low in relation to the amount of RBC
(0/ and +/) the Hb A2 level is characteristically elevated and hyperplasia and hemolysis present. The inappropriate reticulo-
cytosis results from the death of the RBC precursors in the bone
marrow (ineffective erythropoiesis). Due to the deficit in
chains, the unpaired excess chains precipitate in the RBC
precursors, forming inclusion bodies. The inclusion bodies
damage the cell membrane, which leads to destruction of the
cell in the bone marrow.10,12
Electrophoresis or HPLC shows that most of the hemoglo-
bin is Hb F, with a slightly increased level of Hb A2. Hb A is
absent or decreased depending on the specific genotype, which
determines whether none (0/0) or only a small amount
(+/+ or 0/+) of chains are produced. Hb A is present only
if a + gene is present. The bone marrow shows marked ery-
throid hyperplasia, with a myeloid-to-erythroid (M:E) ratio of
1:20 (normal M:E ratio is 1.5:1 to 3.3:1). As a result of the
ineffective erythropoiesis and the hemolysis, the haptoglobin
Figure 27-2 Red blood cells (RBCs) from a patient with -thalassemia level is low and the lactate dehydrogenase level is elevated.
minor, showing microcytic, hypochromic RBCs with target cells, other poi- Transfusions are the major therapeutic option for patients
kilocytes, and basophilic stippling (arrow). with thalassemia major and typically are initiated in the first
396 PART IV Hematopathology: Erythrocyte Disorders

A B
Figure 27-3 Bone changes in severe -thalassemia major. A, Skull radiograph demonstrating the typical hair on end appearance. B, Radiograph of
forearm and hand exhibiting a lacy appearance. (A from Beck WS, editor: Hematology, ed 2, Boston, 1976, MIT Press; B from Nathan DG: Thalassemia,
N Engl J Med 286:586-594, 1972. Copyright 1972 Massachusetts Medical Society. All rights reserved; reprinted with permission.)

year of life.21,22 Beginning in the mid-1970s, it became the


practice to give regular transfusions to prevent not only the
anemia, but also the other consequences of the disorder.
The hemoglobin level is usually maintained above 9g/dL.22
Such transfusion regimens are termed hypertransfusion and are
used to correct the anemia and to suppress the marked eryth-
ropoiesis. With erythropoiesis suppressed, the marked marrow
expansion does not occur, and therefore the bone changes do
not take place. Children receiving this therapy do not develop
hepatosplenomegaly and have much-improved growth and
development.23 The transfusion regimens, however, lead to an
excess iron burden. Because there is no effective physiologic
A pathway for iron excretion in the body, the iron contained in
the transfused RBCs accumulates in the body. This iron is
stored in organs outside the bone marrow (e.g., liver, heart,
pancreas), which results in organ damage. The accumulation
of iron in the liver leads to cirrhosis, and the deposition of iron
in the heart leads to cardiac dysfunction and arrhythmias. In
the past, with transfusion therapy alone, thalassemic patients
died in their teens, typically from cardiac failure. Now patients
undergo iron chelation therapy with the transfusion therapy.
Two drugs, deferoxamine and deferasirox, are now available
that bind excess iron so that it can be excreted in the urine.
Treatment with these agents has been able to prevent iron
accumulation and the subsequent complications of iron
B overload, helping to extend life expectancy of patients with
Figure 27-4 Red blood cells from a patient with -thalassemia major. -thalassemia major well into the thirties (Chapter 19).22,24
Note basophilic stippling, microcytosis, hypochromia, target cells, nucleated Bone marrow transplantation is the only curative therapy
red blood cells, and red cell fragments. A, 500. B, 1000. (Adapted from for thalassemia major.25-27 The average event-free survival rate
Carr JH, Rodak BF: Clinical hematology atlas, ed 2, Philadelphia, 2004, is greater than 80% in children with good risk profiles.27 Patients
Saunders.) with good chelation therapy regimens, no hepatomegaly, and
CHAPTER 27 Thalassemias 397
no portal fibrosis have the highest success rate. Bone marrow production of more chains and greater imbalance of the -
transplantation is not available to all patients, because the chain ratio.33
donor must be an immunologic match. Preferably, this match Because of the heterogeneity of this clinical syndrome, the
would be in a sibling, but there is only a 25% chance that a laboratory and clinical features vary. The RBC morphology of
sibling will have the same immunologic genotype. Unrelated thalassemia intermedia is similar to that previously described
donors can be used as well, but finding an immunologic match for thalassemia major. The degree of anemia and jaundice
in an unrelated donor is less likely. Medications are being evalu- varies depending on the severity of the genetic defect. Because
ated that cause the gene to switch on so that the patients of the presence of splenomegaly, the platelet and neutrophil
RBCs produce higher levels of Hb F.28 Of these various agents, counts may be low. The clinical course varies from minimal
hydroxyurea appears the most successful, as it has been shown symptoms (despite moderately severe anemia) to severe exer-
to increase the Hb F levels enough to eliminate transfusion cise intolerance and pathologic fractures.12 Patients with thalas-
requirements for patients with thalassemia major.29 Not all semia intermedia may also have problems of iron overload
patients appear to respond to hydroxyurea, however, and recent even though they do not receive transfusions. The markedly
studies are searching for genetic factors that will predict accelerated, although ineffective, erythropoiesis, with the
response.30 Ideally, in the future, gene therapy will be able to resulting increase in plasma iron turnover, provokes an increase
correct the genetic defect.31 in gastrointestinal iron absorption.34 Cardiac and endocrine
complications, however, present 10 to 20 years later in thalas-
-Thalassemia Intermedia semia intermedia patients than in patients who receive regular
Thalassemia intermedia is a term used to describe anemia that transfusions.
is more severe than -thalassemia minor but does not require
regular transfusions to maintain hemoglobin level and quality Other Thalassemias Caused by Defects in
of life.12,13 Although patients with thalassemia intermedia typi- the -Globin Gene Cluster
cally maintain a hemoglobin level greater than 7g/dL, it is the Other thalassemias may be caused by deletion, inactivation, or
clinical features rather than the hemoglobin level that deter- fusion of a combination of genes of the -globin gene cluster,
mine the diagnosis.12 In these patients the imbalance in the such as hereditary persistence of fetal hemoglobin (HPFH),
and chain synthesis falls between that observed in 0-thalassemia, and Hb Lepore thalassemia.12,35,36
-thalassemia minor and -thalassemia major, and the general
clinical phenotype falls between the extremes of transfusion- Thalassemias with Increased Levels of
dependent thalassemia major and asymptomatic thalassemia Fetal Hemoglobin
minor. The genotypes of thalassemia intermedia show great HPFH and 0-thalassemia are closely related, heterogeneous
heterogeneity. These patients can be homozygous for muta- conditions that are characterized by continued synthesis
tions that cause a mild decrease in -globin expression. Con- of increased levels of Hb F in adult life. These conditions
versely, they may be compound heterozygous, with one gene have similarities, but can be differentiated by the clinical pre-
causing a mild decrease in -globin production and the other sentation, hemoglobin level, MCV, and amount of Hb F
causing a marked reduction in -globin production.13 In some produced.10,37
instances, only one of the genes carries a mutation, but it is In HPFH, the -globin gene cluster typically contains a
severe enough to cause a significant anemia. Many of the thal- deletion in the region that leads to the increased production
assemia intermedia phenotypes are generated from the coin- of Hb F. However, there are also HPFH conditions that have
heritance of one or two abnormal -globin genes with another intact -globin gene clusters with nondeletional mutations in
hemoglobin defect, such as abnormal genes or unstable the promoter region of the genes that lead to the increased
hemoglobins.12 The coinheritance of -thalassemia may permit Hb F production.10,12,38 Because individuals with these muta-
homozygotes with more severe -thalassemia mutations to tions are characteristically asymptomatic, this condition is of
remain transfusion independent, because the - chain ratio little significance except when it interacts with other forms of
is more balanced so there are fewer free chains to precipitate thalassemia or structural hemoglobin variants, such as Hb S.
and cause hemolysis.32 When genetic changes that increase The additional chains produced are able to replace the
-globin chain production are combined with -thalassemia missing chains and help to restore the balance of and
mutations with minimal -globin chain production, a less non- chains ( or ), which raises the hemoglobin level. Sig-
severe anemia develops. The increase in Hb F production (22) nificant variation is seen in HPFH heterozygotes, but these
helps to compensate for the reduction in Hb A, while helping patients typically are asymptomatic with a normal MCV and
to correct the / balance. Examples of these situations are the Hb F levels of 15% to 30%. Homozygotes for HPFH are also
deletion forms of 0-thalassemia. Individuals homozygous asymptomatic, have a normal to slightly increased hemoglobin
for these mutations, or compound heterozygotes for 0- level, and have slightly hypochromic, microcytic RBCs with
thalassemia and a -thalassemia mutation, have thalassemia 100% Hb F. The increase in hemoglobin observed in some
intermedia with increased chain and Hb F synthesis.13 Coin- patients is likely a response to the slight hypoxia induced by
heritance of a triplicated -globin locus () (see section the higher oxygen affinity of Hb F compared with Hb A.35
on -thalassemia) is also a cause of thalassemia intermedia When assessed using the Kleihauer-Betke acid elution stain
in individuals heterozygous for -thalassemia due to the (discussed later), the distribution of Hb F in HPFH is usually
398 PART IV Hematopathology: Erythrocyte Disorders

pancellular, but can be heterocellular depending on the variant. Nondeletional mutations (mostly point mutations) also occur
In contrast, the Hb F distribution in the other -globin gene in -thalassemia, but are uncommon.10,40,41 The extent of
cluster thalassemias is heterocellular.35 decreased production of the chain depends on what the
The 0-thalassemias are also characterized by deletions in specific mutation is, how many genes are affected, and
the and genes and an increase in Hb F in adult life. Non- whether an 2 or 1 gene is affected. The 2 gene produces
deletion types have also been described.12 In this condition, approximately 75% of the -globin chains in normal RBCs, so
however, the increase in production of the chains is not suf- mutations in the 2 gene generally cause more severe anemia
ficient to completely restore the balance between the and than mutations affecting the 1 gene10,41 The notation for the
non- chains. Heterozygous 0-thalassemia patients have a normal gene complex or haplotype is , which signifies
slight decrease in hemoglobin levels and hypochromic, micro- the two normal genes (2 and 1) on one chromosome 16. A
cytic RBCs with Hb F levels of 5% to 20%. Homozygous 0- normal genotype is /.
thalassemia individuals have hypochromic, microcytic RBCs, The -thalassemias can thus be divided into two haplo-
100% Hb F, and a -thalassemia intermedia phenotype.10,12,35 types: 0-thalassemia and +-thalassemia. In the 0-thalassemia
haplotype (originally named -thal-1), no chains are pro-
Hemoglobin Lepore Thalassemia duced from the -globin gene complex on one chromosome
A rare class of -thalassemias result in a structural hemoglobin due to a deletion of both genes. The designation, , is used
variant called Lepore that has, as the non-globin chain, a for the 0-thalassemia haplotype.10,40 There are 29 known
fusion chain that is composed of the first 50 to 80 amino acid mutations that produce the 0-thalassemia haplotype and
residues of the chain and the last 60 to 90 residues of the involve deletion of both of the genes or the entire -globin
normal C-terminal amino acid sequence of the chain.10,12 gene cluster (including the - and -globin genes) on one
This fusion event occurs during meiosis, from nonhomologous chromosome.10 The 0 haplotype () is found in up to 4%
crossing over between the locus on one chromosome and the of the population in Southeast Asia, is found less frequently in
locus on the other chromosome. Patients with the Lepore the Mediterranean area, and occurs sporadically in other parts
globin chain have reduced production of the fusion globin of the world.5
chain, because the transcriptional control is from the chain The +-thalassemia haplotype (originally named -thal-2)
gene promoter, which is much less active than the chain has decreased chain production from the -globin gene
promoter (Hb A2 [22] levels are normally only 0% to 3.5% complex on one chromosome due to deletional or nondele-
throughout life). In heterozygotes for Hb Lepore the anemia is tional mutations of one of the two genes.41 The designation,
similar to -thalassemia minor, whereas in homozygotes the , is used for the deletional mutations while the designation,
disorder manifests more like -thalassemia major. Conversely, T, is used for the nondeletional mutations. The deletional
in anti-Lepore (the reciprocal fusion on the other chromo- + haplotype () is by far the most common of the thalas-
some), the gene locus is intact, so normal production of the semia haplotypes. It is widely distributed throughout the thal-
chain occurs.10 assemia belt and central Africa (see Figure 26-3), with a carrier
frequency reaching 50% to 80% in some regions of Saudi
Screening for -Thalassemia Minor Arabia, India, Southeast Asia, and Africa.5 The haplotype is
Because of the high frequency of thalassemia in several areas also found in about 30% of African Americans.41 The nonde-
of the world, screening has become an important global health letional + haplotype (T) is relatively uncommon.10,41 More
issue.5 Mass screening programs in Italy and Greece combined than 30 different mutations are known, the majority of which
with prenatal diagnosis have led to a significant reduction are point mutations that affect the predominant 2 gene.10,41
in the number of children born with -thalassemia major.10 The T haplotype produces unstable -globin chains or fewer
Potential carriers of these disorders can be identified by mea- -globin chains than in the haplotype, and generally results
suring the hemoglobin, MCV, Hb A2 and Hb F concentrations, in a more severe anemia.10,40,41
and osmotic fragility.10,39 Other causes of microcytic anemias, One of the most common nondeletional gene mutations
such as iron deficiency, need to be ruled out. When parents are is Constant Spring (2142StopGln), also called CS, haplotype,
identified as potential carriers, DNA can be analyzed by poly- CS.40 It is the result of a point mutation in the 2 gene that
merase chain reaction and molecular hybridization techniques changes the stop codon at 142 to a glutamine codon.10,41 As a
or DNA sequencing for the presence of thalassemia mutations. result of the mutation, additional bases are added to the end
When the parents defect is known, more focused (and less of the mRNA during transcription until the next stop codon is
costly) DNA analyses can be performed in the antenatal reached. The elongated mRNA is very unstable and produces
period.39 only a small amount of the CS chain.40,41 The CS chains, with
an additional 31 amino acids added to the C-terminal end,
combine with chains to form Hb Constant Spring, but the
-THALASSEMIAS
incorporation of a longer chain makes the tetramer unsta-
In contrast to -thalassemia, in which point mutations in the ble.40 Because of the instability of both the mRNA and the Hb
-globin gene cluster are the most common type of mutation, tetramer, the circulating level of Hb Constant Spring is very
in -thalassemia large deletions in the -globin genes (called low.10,40,41 Consequently, hemoglobin Constant Spring is dif-
deletional mutations) are the predominant genetic defect.10,12 ficult to detect by cellulose acetate hemoglobin electrophoresis
CHAPTER 27 Thalassemias 399
at an alkaline pH, and when present, is visualized as a faint, nondeletional + mutation in one -globin gene (T/) also
slow-moving band near the point of origin.10 results in the silent carrier state. In the heterozygous mutation,
CS/, Hb Constant Spring is less than 1% of the total
Clinical Syndromes of -Thalassemia hemoglobin.10
Four clinical syndromes are present in -thalassemia depend-
ing on the gene number, cis or trans pairing, and the amount -Thalassemia Minor
of chains produced.10,12 The four syndromes are silent carrier, -Thalassemia minor can be caused by the homozygous +
-thalassemia minor, Hb H disease, and Hb Bart hydrops (/) or heterozygous 0 (/) forms.10,12 This syndrome
fetalis syndrome10 (Table 27-4). is asymptomatic and characterized by a mild anemia (typical
hemoglobin level is 12 to 13g/dL) with microcytic, hypochro-
Silent Carrier State mic RBCs. At birth, the proportion of Hb Bart is in the range
The deletion of one -globin gene leaving three functional of 5% to 15%.10 Interestingly, in adults the production of
-globin genes (/) is the major cause of the silent carrier and chains is balanced, so Hb H (4) is not usually present.
state. The - chain ratio is nearly normal, and no hematologic Homozygosity for nondeletional mutations in both 2 genes
abnormalities are present.10,12 Because one chain gene is (T/T) produces a mild to moderate hemolytic anemia
absent, there is a slight decrease in chain production. There often with jaundice and hepatosplenomegaly.10,40 In the homo-
is a slight excess of chains at birth that form tetramers of Hb zygous mutation, CS/CS, Hb Constant Spring is 5% to 6%
Bart (4) in the range of 1% to 2%. There is no reliable way to of the total hemoglobin.10,40
diagnose silent carrier state other than genetic analysis. A
Hemoglobin H Disease
Hb H disease is usually caused by the presence of only one
gene producing chains (/).10,12 This genetic abnormality
TABLE 27-4 Some Clinical Syndromes of is particularly common in Asians because of the prevalence of
-Thalassemia the 0 gene haplotype (). It is characterized by the accumula-
Hb Bart Hb H Hb Constant tion of excess unpaired chains that form tetramers of Hb H.
Genotype Hb A (in Newborn) (in Adult) Spring In the newborn, Hb Bart comprises 10% to 40% of the hemo-
globin, with the remainder being Hb F and Hb A. After the
Normal (Normal Hematologic Parameters)
to switch, Hb H replaces most of the Hb Bart, so that Hb H
/ N 0 0 0
is in the range of 5% to 40%, with a reduced amount of Hb
A2, traces of Hb Bart, and the remainder Hb A.10,41 The nonde-
Silent Carrier State (Normal Hematologic
letional + haplotypes, when combined with the 0 haplotype
Parameters)
(/T), generally produces a more severe Hb H disease with
/ N 1-2% 0 0
a higher level of Hb H than the 0 interaction with the dele-
CS/ N 1-3% 0 <1%
tional + haplotype (/).10,40,41 Hb H-Hb Constant Spring
(/CS) is an example.40
-Thalassemia Minor (Asymptomatic; Mild
Hb H disease is characterized by a mild to moderate, chronic
Hemolytic Anemia; Microcytic, Hypochromic)
hemolytic anemia with hemoglobin concentrations averaging
/ Sl 5-15% 0 0
7 to 10g/dL, and reticulocyte counts of 5% to 10%, although
/ Sl 5-15% 0 0
a wide variability in clinical and laboratory findings exist.12,41,42
CS/CS* Sl 5-15% 0 <6%
The bone marrow exhibits erythroid hyperplasia, and the
spleen is usually enlarged. As in most chronic hemolytic states,
Hb H Disease (Moderate Hemolytic Anemia with
infection, pregnancy, or exposure to oxidative drugs may cause
Few Transfusion Requirements; Microcytic,
a hemolytic crisis.
Hypochromic)
Hemolytic crises often lead to the detection of the disease,
/ 10-40% 5-40% 0
because individuals with Hb H disease may otherwise be
/CS <1%
asymptomatic. The RBCs are microcytic and hypochromic with
marked poikilocytosis, including target cells and bizarre shapes.
Hb Bart Hydrops Fetalis Syndrome (Lethal: Hb H is vulnerable to oxidation and gradually precipitates in
Infants Stillborn or Die within Hours of Birth; the circulating RBCs to form inclusion bodies of denatured
Severe Anemia) hemoglobin.42 These inclusions alter the shape and viscoelastic
/ 0 80% (with 5-20% NA 0
properties of the RBCs, contributing to the decreased RBC
Hb Portland)
survival. Splenectomy is beneficial in patients with markedly
*CS/CS genotype results in mild to moderate hemolytic anemia with jaundice and enlarged spleens.41 When incubated with brilliant cresyl blue
hepatosplenomegaly. or new methylene blue, RBCs with Hb H display fine, evenly

/CS genotype results in Hb H disease that is more severe than the / distributed, granular inclusions. These inclusions are typically
genotype.
, Decreased; , Increased more than /; 0, absent; <, less than; CS, Constant removed as the RBC passes through the spleen. Before splenec-
Spring; Hb, hemoglobin; N, normal; NA, not applicable. tomy, only a portion of the cells have this characteristic, but
400 PART IV Hematopathology: Erythrocyte Disorders

Hydropic pregnancies are hazardous to the mother, result-


ing in toxemia and severe postpartum hemorrhage.14 Hydropic
changes are detected in mid-gestation by means of ultrasound
testing.45 If both parents carry one 0-thalassemia haplotype
(/), prenatal diagnosis of homozygosity can be made
by various DNA analyses of tissues obtained from chorionic
villus sampling, amniotic fluid, or cordocentesis.14 Absence
of the -globin genes establishes the diagnosis. Early ter
mination of the pregnancy prevents the serious maternal
complications.

THALASSEMIA ASSOCIATED WITH


STRUCTURAL HEMOGLOBIN VARIANTS
Figure 27-5 Red blood cells from a patient with hemoglobin H disease,
incubated with brilliant cresyl blue, which have acquired fine, evenly Hemoglobin SThalassemia
dispersed granular inclusions. (From the American Society for Hematology Sickle cell anemia (Hb SS)-thalassemia is a genetic abnor-
slide bank.) mality due to the coinheritance of two abnormal genes for Hb
S (at the gene loci) and an -thalassemia haplotype (at the
gene loci). Hb SS+-thalassemia is fairly common, because
after the spleen is removed, most of the RBCs are full of these the genes for Hb S and the +-thalassemia haplotype, ,
inclusions. These cells are often described as golf balls or are common in populations of African ancestry. Individuals
raspberries (Figure 27-5). with Hb SS+-thalassemia have a milder anemia with higher
Two distinct conditions associated with Hb H disease and hemoglobin levels and lower reticulocyte counts than those
mental retardation have been described: ATR-16 syndrome and with sickle cell anemia.46 In one study, Hb SS individuals with
ATR-X syndrome. Chromosomal rearrangements that alter the genotypes, /, /, and /, had average hemo-
the short arm of chromosome 16 (which includes the - globin levels of 8.4, 9.0, and 9.5g/dL, respectively, and reticu-
globin gene cluster) seem to lead to congenital abnormalities, locyte counts of 10.8%, 8.8%, and 6.9%, respectively.46
mental retardation, and Hb H disease, and have been desig- Hb S-thalassemia is a compound heterozygous condition
nated the ATR-16 syndrome.41,43 The genes responsible for the that results from the inheritance of a -thalassemia gene
mental retardation are not yet known. The X-linked disorder from one parent and an Hb S gene from the other. This syn-
(ATR-X syndrome) is a result of mutations of the ATRX gene drome has been reported in the populations of Africa, the
located on the X chromosome.41 This ATR-X syndrome is more Mediterranean area, the Middle East, and India.10 The clinical
common than ATR-16 and is characterized by pronounced expression of Hb S-thalassemia depends on the type of
intellectual and physical deficits. The function of the ATRX -thalassemia mutation inherited.10,12 Individuals with Hb
protein is not well known, but it may play a role in genetic S+-thalassemia produce only a small amount of normal
expression of various proteins involved in RBC development.41 chains, so the clinical syndrome is similar to that of sickle cell
An acquired Hb H disease has also been associated with myelo- anemia. Patients have mostly Hb S with slightly elevated Hb
dysplastic disorders and chronic myelomonocytic leukemia.44 A2 and variable amounts of Hb F and Hb A, depending on the
specific abnormal + gene inherited. These patients can be
Hb Bart Hydrops Fetalis Syndrome distinguished from those with sickle cell anemia by the pres-
Homozygous 0-thalassemia (/) results in the absence of ence of microcytosis, splenomegaly, and some Hb A.
all chain synthesis and is incompatible with life.12,14 The The interaction of silent-thalassemia (in which chains are
infant is born with hydrops fetalis, and edema in the fetal produced at mildly reduced levels) and Hb S results in a condi-
subcutaneous tissues is a result of the severe anemia. Hb Bart tion that may be slightly more severe than sickle cell trait.
(4) is the predominant hemoglobin, along with 5% to 20% Typically, there is mild hemolytic anemia with splenomegaly.
Hb Portland (22) and traces of Hb H.14,42 Hb Bart has high These patients can be distinguished from patients with sickle
oxygen affinity; it does not deliver oxygen to the tissues. The cell trait by the presence of microcytosis and splenomegaly.
fetus survives until the third trimester because of Hb Portland, Hemoglobin electrophoresis or HPLC confirms this condition
but this hemoglobin cannot support the later stages of fetal when the quantity of Hb S exceeds that of Hb A. In sickle cell
growth, and the affected fetus is anoxic. The fetus is delivered trait, Hb A is the predominant hemoglobin.
prematurely and is stillborn or dies shortly after birth. In addi- The combination of 0-thalassemia and Hb S produces a
tion to anemia, edema, and ascites, the fetus has gross hepato- phenotype similar to sickle cell anemia with a similar inci-
splenomegaly and cardiomegaly. At delivery, there is a severe dence of stroke and a similar life expectancy.47 Both conditions
microcytic, hypochromic anemia with numerous nucleated lack Hb A and produce severe pain crises as the predominant
RBCs in the peripheral blood. The bone marrow cavity is symptom. Typically, the microcytosis and elevated Hb A2
expanded and marked erythroid hyperplasia is present, as well level in Hb S0-thalassemia distinguish it from sickle cell
as extramedullary erythropoiesis. anemia.
CHAPTER 27 Thalassemias 401

TABLE 27-5 Thalassemias Associated with Structural Hemoglobin (Hb) Variants (Compound Heterozygotes)
Genotype Hb A Hb A2 Hb F Other Hb RBC Morphology Clinical Manifestations* Treatment
+
Hb S -thalassemia N to Hb S > Hb A Microcytes, sickle Ranges from mild to severe Ranges from no treatment
Hb S0-thalassemia 0 N to Hb S cells, target anemia with recurrent to transfusion support
cells vasoocclusive crises and pain control
Hb C+-thalassemia
Hb C > Hb A Microcytes, Hb C Ranges from moderate to Usually no treatment
Hb C0-thalassemia 0
Hb C crystals, target severe anemia needed
cells
Hb E+-thalassemia
Hb E > Hb A Microcytes, target Ranges from mild to severe Ranges from no treatment
Hb E0-thalassemia 0
Hb E cells anemia with transfusion to transfusion support
dependency

, Increased; , decreased; 0, absent; N, normal; RBC, red blood cell.


*Clinical manifestations depend on the amount of Hb A produced; compound heterozygotes with the 0 gene have more severe symptoms.

Not all methods can quantitate Hb A2 in the presence of the abnormal Hb. High-performance liquid chromatography can separate Hb A2 from Hb C; capillary electrophoresis can
separate Hb A2 from Hb E.

Hemoglobin CThalassemia count is disproportionately high relative to the degree of


Hb C-thalassemia produces moderately severe hemolysis, anemia, which generates a very low MCV. The mean cell
splenomegaly, hypochromia, microcytosis, and numerous hemoglobin concentration (MCHC) is slightly decreased. The
target cells. The hemoglobin electrophoresis pattern varies RBC distribution width (RDW) is elevated (reflecting anisocy-
depending on the type of -thalassemia gene defect, with tosis) in untreated -thalassemia major, but is often normal
higher Hb C concentrations in patients when there is minimal in -thalassemia minor. On a Wright-stained peripheral blood
or no -globin production.10 film, the RBCs are typically microcytic and hypochromic
except in the silent carrier phenotypes, in which the RBCs
Hemoglobin EThalassemia appear normal. In -thalassemia minor and Hb H disease, the
Hb E-thalassemia is a significant concern in Southeast Asia cells are microcytic with slight to moderate poikilocytosis.
owing to the high prevalence of both genetic mutations. In heterozygous 0-thalassemia, there is mild hypochromia
Because Hb E is synthesized at a reduced rate, the disorder and microcytosis but less poikilocytosis. In homozygous and
clinically appears as a mild -thalassemia in the homozygous compound heterozygous -thalassemia, extreme poikilocyto-
state. When the mutations are coinherited in the compound sis may be present, including target cells and elliptocytes, in
heterozygous state, there may be a marked reduction of chain addition to polychromasia, basophilic stippling, and nucle-
production. The clinical picture ranges from thalassemia inter- ated RBCs.
media to a transfusion-dependent condition that is indistin-
guishable from homozygous -thalassemia.10 Reticulocyte Count
Table 27-5 summarizes some compound heterozygous The reticulocyte count is elevated, which indicates that the
states of -thalassemia combined with a structural hemoglobin bone marrow is responding to a hemolytic process. In Hb H
defect. disease, the typical reticulocyte count is 5% to 10%.10 In homo-
zygous -thalassemia, it is 2% to 8%, disproportionately low
relative to the degree of anemia. An inadequate reticulocytosis
DIAGNOSIS OF THALASSEMIA
reflects ineffective erythropoiesis.10,12
History and Physical Examination
Individual and family histories are paramount in the diagnosis Bone Marrow Examination
of thalassemia. The race or ethnic background of the individual The bone marrow in untreated -thalassemia major is
should be investigated because of the increased frequency of markedly hypercellular with extreme erythroid hyperplasia.
various abnormal genes in certain populations. In the clinical Extramedullary hematopoiesis is prominent. In Hb H disease
examination, pallor indicating anemia, jaundice indicating the bone marrow shows less erythroid hyperplasia. Only slight
hemolysis, splenomegaly caused by pooling of the abnormal erythroid hyperplasia is seen in thalassemia minor.
cells, and skeletal deformities, especially marked in untreated
-thalassemia major as a result of expansion of the bone marrow Osmotic Fragility Test
cavities, are findings that suggest thalassemia (Figure 27-6).10 The microcytic, hypochromic RBCs of the thalassemias have a
decreased osmotic fragility. This characteristic has been used
Laboratory Findings to screen for the thalassemia carrier state in populations in
Complete Blood Count which thalassemia is common, and it has been found to
Most thalassemias are microcytic and hypochromic. The have an excellent negative predictive value (greater than
hemoglobin and hematocrit (Hct) are decreased, but the RBC 99%).48 Because iron deficiency also leads to decreased osmotic
402 PART IV Hematopathology: Erythrocyte Disorders

Figure 27-6 Flow chart showing the approach to the diagnosis of the thalassemia syndromes. Hb, Hemoglobin; MCH, mean cell hemoglobin; MCV, mean
cell volume; retics, reticulocytes, RBC, red blood cell. (From Weatherall DJ: The thalassemias. In Beutler E, Lichtman MA, Coller BS, etal, editors: Williams
hematology, ed 6, New York, 2001, McGraw-Hill, p 533.)

N SCT SCA S-T S-0T T 0T TT 0T HPFH


A
F
S
C, A2

Figure 27-7 Relative electrophoretic mobilities on cellulose acetate (pH 8.4) of various hemoglobins (Hbs) important in the diagnosis of thalassemia syn-
dromes and hemoglobinopathies. 0T, 0-thalassemia major, 0/0 (no Hb A, increased Hb F, slight increase in Hb A2); +T, +-thalassemia major, +/+
(decreased Hb A, increased Hb F, slight increase in Hb A2); TT, -thalassemia minor (slight decrease in Hb A, increased Hb A2, some Hb F); 0T, 0-
thalassemia, homozygous, 0/0 (100% Hb F); HPFH, hereditary persistence of fetal hemoglobin, heterozygous (mostly Hb A, some Hb F, no Hb A2); N,
normal; SCA, sickle cell anemia (no Hb A, mostly Hb S, increased Hb F, normal Hb A2); SCT, sickle cell trait (Hb A > Hb S, normal Hb A2 and F); S-0T, sickle
cell0-thalassemia (no Hb A, increased Hb A2 and F, mostly Hb S); S- +T, sickle cell+-thalassemia (Hb A < Hb S, increased Hb A2 and F).

fragility, the test does not differentiate between these two Electrophoresis
conditions. Hemoglobin electrophoresis on cellulose acetate at alkaline
pH has classically been the major diagnostic tool used to
Supravital Staining detect and identify thalassemia. This technique is able to
In -thalassemia minor, Hb H disease, and silent carrier differentiate hemoglobin variants, such as Hb E, Hb Lepore,
-thalassemia, the brilliant cresyl blue or new methylene blue and Hb Constant Spring,50 and is able to distinguish the
test may be used to induce precipitation of the intrinsically fast-moving Hb H and Hb Bart.51 Electrophoresis is used
unstable Hb H.49 Hb H inclusions (denatured -globin chains) to screen for the elevation in Hb A2 associated with -
typically appear as small, multiple, irregularly shaped greenish thalassemia minor, although specific tests for quantitation of
blue bodies uniformly distributed throughout the RBC. They Hb A2 are more accurate.50 An increase in Hb F can be detected
produce a pitted pattern on the RBCs similar to the pattern of on electrophoresis, which is important in 0-thalassemia,
a golf ball or raspberry (see Figure 27-5). In Hb H disease, HPFH, and other -thalassemia variants (Figure 27-7). Hb F
almost all RBCs contain these Hb H inclusions. In -thalassemia should be quantitated by other, more accurate methods,
minor, only a few cells may contain these inclusions, and in however. Citrate agar gel electrophoresis performed at an
silent carrier -thalassemia, only a rare cell does. These inclu- acid pH differentiates Hb S from Hb D and Hb G, and Hb
sions appear different from Heinz bodies, which are larger and C from Hb E and Hb O (see Figure 26-7).52 Emerging tech-
fewer in number and most often appear eccentrically along the nologies such as isoelectric focusing, capillary electrophoresis,
membrane of the RBC. This test is very sensitive in detecting and HPLC have expanded upon or replaced hemoglobin
Hb H in the -thalassemia conditions.49 electrophoresis.51,52
CHAPTER 27 Thalassemias 403
Hemoglobin A2 and Hemoglobin F Quantitation DNA Analysis. Molecular techniques identify globin gene
Quantitation of Hemoglobin A2 . Hb A2 is separated by mutations.39 With these detailed analyses, gene frequency and
column chromatography from the other hemoglobins and geographic distribution can now be determined. Current
measured. Several techniques are available to quantitate Hb A2, methods include polymerase chain reaction, real-time poly-
but automated HPLC is most commonly used.50 HPLC may merase chain reaction, signal amplification systems, and DNA
also be used to simultaneously measure Hb F. This technique sequencing.39 Many new and rare mutations have been identi-
is unable to differentiate Hb E from Hb A2, however, so its use fied. Molecular techniques are used to aid diagnosis in difficult
is limited when Hb E is present.50 The accuracy of HPLC is cases and, more importantly, to provide information for
diminished in sickle cell trait, because Hb A2 can be slightly genetic counseling. They also can be used in antenatal diagno-
elevated in association with Hb S. Using improved techniques sis, especially if the parents genetic mutations are known.39
and complementing HPLC results with capillary electrophore-
sis, however, have minimized these concerns.53 Indirect Serum Bilirubin Level. The serum level of indi-
rect bilirubin is increased in thalassemias, which confirms the
Quantitation of Hemoglobin F. Hb F is increased in hemolytic component of this disorder. The increase is mild in
homozygous 0-thalassemia, +-thalassemia, 0-thalassemia, thalassemia minor but moderate in thalassemia intermedia
Hb Lepore thalassemia, and HPFH. A moderate or slight eleva- and Hb H disease. Thalassemia major also shows bilirubine-
tion in Hb F can be seen in -thalassemia minor. mia; however, the elevation is mild because of the inability to
The classic alkali denaturation test is accurate and precise for produce hemoglobin.
the quantitation of Hb F in the 0.2% to 50% range.50 Most
human hemoglobins are denatured on exposure to a strong Differential Diagnosis of Thalassemia Minor
alkali, but Hb F is not. The Hb F can be separated and its con- and Iron Deficiency Anemia
centration compared with that of other hemoglobins. Consis- The RBCs in thalassemia minor are microcytic and hypochro-
tent methodology is required to ensure accurate results.50 As mic, and this disease must be differentiated from iron defi-
with Hb A2 quantitation, HPLC is now often used for quantita- ciency anemia and other microcytic, hypochromic anemias.
tive measurement of Hb F. The differential diagnosis for microcytic, hypochromic anemias
In the Kleihauer-Betke acid elution slide test, peripheral is relatively limited (see Table 19-1). Differentiating thalasse-
blood films are immersed in an acid buffer. Adult hemoglobin mia minor from iron deficiency is important to avoid unneces-
is eluted from the RBCs, whereas fetal hemoglobin resists acid sary tests or treatments. An incorrect presumption that a patient
elution and remains in the cell. When the cells are subse- has iron deficiency may lead to inappropriate iron therapy or
quently stained, RBCs containing Hb F will take up the to unnecessary diagnostic procedures such as colonoscopy to
stain, whereas RBCs containing only adult hemoglobin will identify a source of blood loss.
appear as ghosts. This test determines if the Hb F distribution Clinical history is crucial. A family history of thalassemia
in RBCs is pancellular (most HPFH cases) or heterocellular raises the suspicion for this diagnosis. A history of previously
(-globin gene cluster thalassemias and some HPFH cases).35 normal hemoglobin levels and RBC indices, significant bleed-
The Kleihauer-Betke slide test is also used to estimate the ing, or pica leads to the diagnosis of iron deficiency.55 Pica
volume of fetal-maternal hemorrhage to determine if an means cravings for nonfood items such as clay, dirt, or starch.
increased dose of Rh immune globulin is needed for an The most common pica symptom in the United States is pago-
Rh-negative mother who delivers an Rh-positive baby. Because phagia, the craving to chew on ice.55
the Kleihauer-Betke slide test is cumbersome to perform and Iron deficiency and -thalassemia minor are best differenti-
results are difficult to replicate, flow cytometry is becoming the ated using serum ferritin level, serum iron level, total iron-
standard test to measure fetal-maternal hemorrhage quickly binding capacity, transferrin saturation, and Hb A2 level, along
and accurately.54 with a complete blood count (CBC) and examination of a
peripheral blood film.55,56 Additional testing may also include
soluble transferrin receptor and zinc protoporphyrin levels for
Other Special Procedures iron deficiency (see Chapter 19).55
Other special procedures such as mass spectrometry and DNA Before evaluating Hb A2 levels for -thalassemia minor, iron
analysis can be used to identify specific genotypes for research deficiency should be ruled out. Low iron levels in patients with
purposes, to differentiate an -thalassemia carrier from an - -thalassemia minor decrease the Hb A2 levels.56 The iron
thalassemia carrier, to identify a silent carrier gene, or to deter- stores need to be replenished before the laboratory analysis for
mine family inheritance patterns with multiple gene mutations. thalassemia is undertaken.
This further testing is done in reference laboratories. A mild erythrocytosis (high RBC count) and marked micro-
cytosis (low MCV) are found more commonly in -thalassemia
Mass Spectometry. Using whole blood, mass spectrom- minor. In iron deficiency anemia, the RBC count and MCV may
etry assesses the difference in mass of the globin chains to be normal or decreased depending on whether the deficiency
detect single amino acid substitutions. This technique typically is developing or longstanding.57-59 The RDW can be normal or
is used in conjunction with electrophoresis or HPLC to identify increased in both -thalassemia minor and iron deficiency
variant hemoglobin molecules.50 anemia with a significant overlap of values; therefore, the
404 PART IV Hematopathology: Erythrocyte Disorders

RDW alone cannot distinguish these conditions.56-58 Various deficiency anemia ranged from 60% to 96% in various studies,
discrimination indices have been proposed to distinguish which leads to a high number of false-negative results. Thus
-thalassemia minor from iron deficiency anemia using a cal- their use for screening is not appropriate.12,55-57 The peripheral
culation based on the RBC count, hemoglobin level, MCV, blood film may demonstrate basophilic stippling in
mean cell hemoglobin (MCH), and/or RDW (such as the -thalassemia minor, which can distinguish it from iron defi-
Mentzer, Green and King, England and Fraser, Shine and Lal, ciency. Because target cells can be found in both conditions,
and Srivastava indices).56-58 Unfortunately, the sensitivity of however, their presence does not help discriminate between
these indices in discriminating -thalassemia minor and iron the two disorders.

SUMMARY
Thalassemias are a group of heterogeneous disorders in which The -thalassemias are divided clinically into silent carrier state,
one or more globin chains are reduced or absent. -thalassemia minor, Hb H disease, and Hb Bart hydrops fetalis.
Thalassemias result in a hypochromic, microcytic anemia due to Silent carrier -thalassemia is a result of the deletion, or rarely a
decreased production of hemoglobin. nondeletional mutation, of one of four genes (/) or
The imbalance of globin chain synthesis causes an excess of the (T/); it is associated with a normal RBC profile and is
normally produced globin chain that damages the RBCs or their asymptomatic.
precursors and results in hemolysis. -Thalassemia minor is a result of the deletion of two genes
0 refers to a thalassemic gene that results in production of no (/ or /) and is clinically similar to -thalassemia minor
chains. except that Hb A2 is not increased.
+ refers to a thalassemic gene that results in production of Homozygosity for nondeletional mutations in both 2 genes
reduced amounts of chains. (T/T) produces a mild to moderate hemolytic anemia, often
-Thalassemia is clinically manifested as silent carrier state, with jaundice and hepatosplenomegaly.
thalassemia minor, thalassemia intermedia, or thalassemia Hb H disease is a result of the deletion of three of the four genes
major. (/); Hb H inclusions (excess chains) precipitate in older
The silent carrier state (silent/) is caused by one mutated gene circulating RBCs, causing a hemolytic anemia. The RBCs are
that produces a small decrease in chains. The blood picture is microcytic and hypochromic, and the disease is clinically similar
completely normal in the silent carrier state. to -thalassemia intermedia.
-Thalassemia minor is a mild, asymptomatic, microcytic, hypo- The genotype (/T) generally produces a more severe Hb H
chromic anemia; it is usually characterized by an elevated Hb A2 disease with a higher level of Hb H than the (/) genotype.
level, which aids in diagnosis. Hb Bart hydrops fetalis syndrome, in which all four of the genes
-Thalassemia major is a severe anemia leading to transfusion are deleted (/) results in severe anemia in utero and is
dependence. incompatible with life.
-Thalassemia intermedia manifests abnormalities with a severity The diagnosis of thalassemia is made from CBC results and RBC
between those of -thalassemia major and -thalassemia minor. morphology, hemoglobin electrophoresis or HPLC, Hb A2 and
Hb Lepore thalassemia, hereditary persistence of fetal hemoglo- Hb F quantitation, and findings on a supravital stain preparation
bin, and 0-thalassemia are other thalassemia syndromes involv- (Hb H disease).
ing the -globin gene cluster on chromosome 11. Thalassemia trait must be differentiated from other microcytic,
The -thalassemias are usually caused by a deletion of one or hypochromic anemias, especially iron deficiency anemia. Iron
both of the genes on chromosome 16, resulting in reduced or studies are important for this differentiation.
absent production of chains.
In -thalassemias, tetramers of chains may precipitate as Hb Now that you have completed this chapter, go back and
Bart in the fetus and newborn, and tetramers of chains may read again the case study at the beginning and respond
precipitate as Hb H in the adult. to the questions presented.

R E V I E W Q UESTIONS
1. The thalassemias are caused by: 2. Thalassemia is more prevalent in individuals from areas
a. Structurally abnormal hemoglobins along the tropics because it confers:
b. Absent or defective synthesis of a polypeptide chain a. Heat resistance to those heterozygous for a
in hemoglobin thalassemia gene
c. Excessive absorption of iron b. Selective advantage against tuberculosis
d. Abnormal or defective porphyrin synthesis c. Selective advantage against malaria
d. Resistance to mosquito bites
CHAPTER 27 Thalassemias 405

3. The hemolytic anemia associated with the thalassemias is 10. When one gene is deleted (/), a patient has:
due to: a. Normal hemoglobin
a. Imbalance of globin chain synthesis b. Mild anemia (hemoglobin range 9 to 11g/dL)
b. Microcytic, hypochromic cells c. Moderate anemia (hemoglobin range 7 to 9gm/dL)
c. Ineffective erythropoiesis caused by immune factors d. Marked anemia requiring regular transfusions
d. Structurally abnormal hemoglobin
11. In which part of the world is the gene mutation causing
4. -Thalassemia minor (heterozygous) usually exhibits: Hb Bart hydrops fetalis (/) most common?
a. Increased Hb Constant Spring a. Northern Africa
b. 50% Hb F b. Mediterranean
c. No Hb A c. Middle East
d. Increased Hb A2 d. Southeast Asia

5. RBC morphologic features in -thalassemia would most 12. The condition Hb S0-thalassemia has a clinical course
likely include: that resembles:
a. Microcytic cells, hypochromic cells, target cells, a. Sickle cell trait
elliptocytes, stippled cells b. Sickle cell anemia
b. Macrocytic cells, ovalocytes, target cells, stippled c. -Thalassemia minor
cells d. -Thalassemia major
c. Microcytic cells, sickle cells
d. Macrocytic cells, hypochromic cells, target cells, 13. Hb H inclusions in a supravital stain preparation appear
stippled cells as:
a. A few large, blue, round bodies in the RBCs with
6. The predominant hemoglobin present in 0-thalassemia aggregated reticulum
major is: b. Uniformly stained blue cytoplasm in the RBC
a. Hb A c. Small, evenly distributed, granular, greenish-blue
b. Hb A2 bodies that make the RBCs resemble golf balls
c. Hb F d. Uniform round bodies that adhere to the RBC
d. Hb C membrane

7. Heterozygous HPFH is characterized by: 14. Which of the following laboratory findings is inconsistent
a. 15% to 30% Hb F with normal RBC morphology with -thalassemia minor?
b. 100% Hb F with slightly hypochromic, microcytic a. A slightly elevated RBC count and marked
cells microcytosis
c. A decreased amount of Hb F with normal RBC b. Target cells and basophilic stippling on the peripheral
morphology blood film
d. 5% to 15% Hb F with hypochromic, macrocytic c. Hemoglobin level of 10 to 13g/dL
cells d. Elevated MCHC and spherocytic RBCs

8. Hb H is composed of: 15. A 4-month-old infant of Asian heritage is seen for a


a. Two and two chains well-baby check. Because of pallor, the physician suspects
b. Two and two chains anemia and orders a CBC. The RBC count is 4.5 109/L,
c. Four chains Hb concentration is 10g/dL, and MCV is 77fL, with
d. Four chains microcytosis, hypochromia, poikilocytosis, and mild poly-
chromasia noted on the peripheral blood film. These find-
9. Hb Bart is composed of: ings lead the physician to suspect:
a. Two and two chains a. -Thalassemia major
b. Two and two chains b. -Thalassemia silent carrier state
c. Four chains c. Iron deficiency anemia
d. Four chains d. Homozygous -thalassemia (/)

REFERENCES
1. Cooley TB, Lee P: A series of cases of splenomegaly in chil- 2. Lichtman MA, Spivak JL, Boxer LA, et al: Hematology: landmark
dren with anemia and peculiar bone changes. Trans Am papers of the twentieth century, San Diego, 2000, Academic
Pediatr Soc 37:29-30, 1925. Press.
406 PART IV Hematopathology: Erythrocyte Disorders

3. Schechter AN: Hemoglobin research and the origins of 24. Cappellini MD, Cohen A, Piga A, et al: A phase 3 study of
molecular medicine. Blood 112:3927-3938, 2008. deferasirox (ICL670), a once-daily oral iron chelator, in
4. Angastiniotis M, Modell B: Global epidemiology of hemoglo- patients with -thalassemia. Blood 107:3455-3462, 2006.
bin disorders. Ann N Y Acad Sci 850:251-269, 1998. 25. Lawson SE, Roberts IA, Amrolia P, et al: Bone marrow
5. Weatherall DJ, Clegg JB: Inherited haemoglobin disorders: an transplantation for beta-thalassaemia major: the UK experi-
increasing global health problem. Bull World Health Organ ence in two paediatric centres. Br J Haematol 120:289-295,
79:704-712, 2001. 2003.
6. Mockenhaupt FP, Ehrhardt S, Gellert S, et al: Alpha(+)- 26. La Nasa G, Argiolu F, Giardini C, et al: Unrelated bone
thalassemia protects African children from severe malaria. marrow transplantation for beta-thalassemia patients: the
Blood 104:2003-2006, 2004. experience of the Italian Bone Marrow Transplant Group. Ann
7. Williams TN, Wambua S, Uyoga S, et al: Both heterozygous N Y Acad Sci 1054:186-195, 2005.
and homozygous + thalassemia protect against severe and 27. Bhatia M, Walters MC: Hematopoietic cell transplantation for
fatal Plasmodium falciparum malaria on the coast of Kenya. thalassemia and sickle cell disease: past, present and future.
Blood 106:368-371, 2005. Bone Marrow Transplant 41:109-117, 2008.
8. Pattanapanyasat K, Yongvanitchit K, Tongtawe P, et al: Impair- 28. Perrine SP: Fetal globin inductioncan it cure thalassemia?
ment of Plasmodium falciparum growth in thalassemic red Hematology Am Soc Hematol Educ Program 38-44, 2005.
blood cells: further evidence by using biotin labeling and 29. Bradai M, Abad MT, Pissard S, et al: Hydroxyurea can elimi-
flow cytometry. Blood 93:3116-3119, 1999. nate transfusion requirements in children with severe beta-
9. Ayi K, Turrini F, Piga A, et al: Enhanced phagocytosis of thalassemia. Blood 102:1529-1530, 2003.
ring-parasitized mutant erythrocytes: a common mechanism 30. Koren A, Levin C, Dgany O, et al: Response to hydroxyurea
that may explain protection against falciparum malaria in therapy in -thalassemia. Am J Hematol 83:366-370, 2008.
sickle trait and beta-thalassemia trait. Blood 104:3364-3371, 31. Puthenveetil G, Scholes J, Carbonell D, et al: Successful cor-
2004. rection of the human beta-thalassemia major phenotype
10. Weatherall DJ: The thalassemias. In Lichtman MA, Beutler E, using a lentiviral vector. Blood 104:3445-3453, 2004.
Kipps TJ, et al, editors: Williams hematology, ed 7, New York, 32. Galanello R, Dessi E, Melis MA, et al: Molecular analysis of
2009, McGraw-Hill, pp 633-666. beta zero-thalassemia intermedia in Sardinia. Blood 74:823-
11. Harju S, McQuenn KJ, Peterson KR: Chromatin structure and 827, 1989.
control of beta-like globin gene switching. Exp Biol Med 33. Oron V, Filon D, Oppenheim A, et al: Severe thalassaemia
227:683-700, 2002. intermedia caused by interaction of homozygosity for alpha-
12. Borgna-Pignatti C, Galanello R: Thalassemias and related dis- globin gene triplication with heterozygosity for beta zero-
orders: quantitative disorders of hemoglobin synthesis. In thalassaemia. Br J Haematol 86:377-379, 1994.
Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes 34. Cazzola M, Beguin Y, Bergamaschi G, et al: Soluble transferrin
clinical hematology, ed 12, Philadelphia, 2009, Wolters receptor as a potential determinant of iron loading in con-
Kluwer Health/Lippincott Williams & Wilkins, pp 1083- genital anaemias due to ineffective erythropoiesis. Br J Hae-
1131. matol 106:752-755, 1999.
13. Galanello R, Cao A: Relationship between genotype and phe- 35. Rochette J, Craig JE, Thein SL: Fetal hemoglobin levels in
notype: thalassemia intermedia. Ann N Y Acad Sci 850:325- adults. Blood Rev 8:213-224, 1994.
333, 1998. 36. Bollekens JA, Forget BG: Delta beta thalassemia and heredi-
14. Chui DHK, Waye JS: Hydrops fetalis caused by -thalassemia: tary persistence of fetal hemoglobin. Hematol Oncol Clin
an emerging health care problem. Blood 91:2213-2222, 1998. North Am 5:399-422, 1991.
15. Thein SL: Genetic insights into the clinical diversity of beta 37. Craig JE, Barnetson RA, Prior J, et al: Rapid detection of dele-
thalassaemia. Br J Haematol 124:264-274, 2004. tions causing delta beta thalassemia and hereditary persis-
16. Thein SL: Pathophysiology of beta thalassemiaa guide to tence of fetal hemoglobin by enzymatic amplification. Blood
molecular therapies. Hematology Am Soc Hematol Educ Program 83:1673-1682, 1994.
31-37, 2005. 38. Tuan D, Feingold E, Newman M, et al: Different 3 end points
17. Murru S, Loudianos G, Deiana M, et al: Molecular character- of deletions causing delta beta-thalassemia and hereditary
ization of beta-thalassemia intermedia in patients of Italian persistence of fetal hemoglobin: implications for the control
descent and identification of three novel beta-thalassemia of gamma-globin gene expression in man. Proc Natl Acad Sci
mutations. Blood 77:1342-1347, 1991. U S A 80:6937-6941, 1983.
18. Gasperini D, Perseu L, Melis MA, et al: Heterozygous beta- 39. Old JM: Screening and genetic diagnosis of haemoglobin
thalassemia with thalassemia intermedia phenotype. Am J disorders. Blood Rev 17:43-53, 2003.
Hematol 57:43-47, 1998. 40. Singsanan S, Fucharoen G, Savongsy O, et al: Molecular char-
19. Gilman JG, Huisman TH, Abels J: Dutch beta 0-thalassaemia: acterization and origins of Hb Constant Spring and Hb Pakse
a 10 kilobase DNA deletion associated with significant in Southeast Asian populations. Ann Hematol 86:665-669,
gamma-chain production. Br J Haematol 56:339-348, 1984. 2007.
20. Oggiano L, Pirastu M, Moi P, et al: Molecular characterization 41. Chui DH, Fucharoen S, Chan V: Hemoglobin H disease: not
of a normal Hb A2 beta-thalassaemia determinant in a necessarily a benign disorder. Blood 101:791-800, 2003.
Sardinian family. Br J Haematol 67:225-229, 1987. 42. Weatherall DJ: The thalassemias. In Stamatoyannopoulos G,
21. Modell B, Khan M, Darlison M: Survival in beta-thalassaemia Majerus PW, Perlmutter RM, et al, editors: The molecular
major in the UK: Data from the UK Thalassaemia Register. basis of blood diseases, ed 3, Philadelphia, 2001, Saunders,
Lancet 355:2051-2052, 2000. pp 183-226.
22. Borgna-Pignatti C, Rugolotto S, De Stefano P, et al: Survival 43. Wilkie AO, Zeitlin HC, Lindenbaum RH, et al: Clinical fea-
and complications in patients with thalassemia major treated tures and molecular analysis of the alpha thalassemia/mental
with transfusion and deferoxamine. Haemotologica 89:1187- retardation syndromes. II. Cases without detectable abnor-
1193, 2004. mality of the alpha globin complex. Am J Hum Genet 46:1127-
23. Olivieri NF, Nathan DG, MacMillan JH, et al: Survival in 1140, 1990.
medically treated patients with homozygous beta-thalassemia. 44. Nelson ME, Thurmes PJ, Hoyer JD, et al: A novel 5
N Engl J Med 331:574-578, 1994. ATRX mutation with splicing consequences in acquired
CHAPTER 27 Thalassemias 407
thalassemia-myelodysplastic syndrome. Haematologica 90: 52. Ou CN, Rognerud CL: Diagnosis of hemoglobinopathies:
1463-1470, 2005. electrophoresis vs. HPLC. Clin Chim Acta 313:187-194,
45. Ko TM, Tseng LH, Hsu PM, et al: Ultrasonographic scanning 2001.
of placental thickness and the prenatal diagnosis of homozy- 53. Keren DF: Comparison of Sebia Capillarys capillary electro-
gous alpha-thalassaemia 1 in the second trimester. Prenat phoresis with the Primus high-pressure liquid chromatogra-
Diagn 15:7-10, 1995. phy in the evaluation of hemoglobinopathies. Am J Clin
46. Steinberg MH, Rosenstock W, Coleman MB, et al: Effects of Pathol 130:824-831, 2008.
thalassemia and microcytosis on the hematologic and vaso- 54. Mundee Y, Bigelow NC, Davis BH, et al: Simplified flow
occlusive severity of sickle cell anemia. Blood 63:1353-1360, cytometric method for fetal hemoglobin containing red
1984. blood cells. Cytometry 42:389-393, 2000.
47. Quinn CT, Rogers ZR, Buchanan GR: Survival of children with 55. Cook JD: Diagnosis and management of iron-deficiency
sickle cell disease. Blood 103:4023-4027, 2004. anaemia. Best Pract Res Clin Haematol 18:319-332, 2005.
48. Sirichotiyakul S, Tantipalakorn C, Sanguansermsri T, et al: 56. Beyan C, Kaptan K, Ifran A: Predictive value of discrimination
Erythrocyte osmotic fragility test for screening of alpha- indices in differential diagnosis of iron deficiency anemia and
thalassemia-1 and beta-thalassemia trait in pregnancy. Int J beta-thalassemia trait. Eur J Haematol 78:524-526, 2007.
Gynaecol Obstet 86:347-350, 2004. 57. Ntaois G, Chatzinikolaou A, Saouli Z, et al: Discrimination
49. Pan LL, Eng HL, Kuo CY, et al: Usefulness of brilliant cresyl indices as screening tests for -thalassemia trait. Ann Hematol
blue staining as an auxiliary method of screening for alpha- 86:487-491, 2007.
thalassemia. J Lab Clin Med 145(2):94-97, 2005. 58. Demir A, Yarali N, Fisgin T, et al: Most reliable indices in
50. Wild BJ, Bain BJ: Detection and quantitation of normal and differentiation between thalassemia trait and iron deficiency
variant haemoglobins: an analytical review. Ann Clin Biochem anemia. Pediatr Int 44:612-616, 2002.
41:355-369, 2004. 59. Means RT, Jr, Glader B: Anemia: general considerations. In
51. Higgins T, Mack M, Khajuria A: Comparison of two methods Greer JP, Foerster J, Rodgers GM, et al, editors: Wintrobes
for the quantification and identification of hemoglobin vari- clinical hematology, ed 12, Philadelphia, 2009, Wolters Kluwer
ants. Clin Biochem 42:701-705, 2009. Health/Lippincott Williams & Wilkins, pp 779-809.
PART V Leukocyte Disorders

28 Nonmalignant Leukocyte Disorders Anne Stiene-Martin

OUTLINE OBJECTIVES
Genetic Leukocyte After the completion of this chapter, the reader will be able to:
Abnormalities
Nuclear Abnormalities 1. Describe the basic genetic defect and the morpho- 8. Discuss at least three other miscellaneous granulo-
Cytoplasmic Abnormalities logic consequences in Pelger-Hut anomaly. cyte disorders.
Functional Abnormalities 2. Indicate with what cell type Pelger-Hut cells might 9. Describe the characteristic macrophage morpho
Reactive or Stress- be confused and discuss what is meant by pseudo- logy associated with the mucopolysaccharidoses,
Related Leukocyte Pelger Hut. Gaucher disease, and Niemann-Pick disease.
Abnormalities 3. Compare and contrast two genetic causes of 10. Describe the basic defect in genetic disorders leading
Quantitative Abnormalities neutrophilic hypersegmentation and indicate what to decreased T-lymphocyte production, decreased
Qualitative (Morphologic) other disorder must be ruled out. B-lymphocyte production, and the combined
Abnormalities 4. Describe the basic genetic defect and the morpho- decrease of T, B, and natural killer lymphocytes.
Infectious Mononucleosis
logic consequences in Alder-Reilly anomaly, Chdiak- 11. Define what is meant by neutrophilia, neutropenia,
Higashi syndrome, and May-Hegglin anomaly. lymphocytosis, lymphocytopenia, monocytosis, mono
5. Indicate how inclusions in Alder-Reilly anomaly and cytopenia, eosinophilia, eosinopenia, and basophilia;
May-Hegglin anomaly might be confused with reac- give some examples of conditions in which each
tive morphology. occurs.
6. Discuss the defect in and functional consequences 12. Describe the nuclear and cytoplasmic alterations
of chronic granulomatous disease. in granulocytes, monocytes, and lymphocytes that
7. Discuss the basic defects in and functional conse- indicate a reaction to infection, inflammation, or
quences of leukocyte adhesion disorders. stress.

CASE STUDIES
After studying the material in this chapter, the reader should be able to respond to the following case studies:

Case 1 3. Do all microorganisms produce equally severe disease in


A 5-year-old boy has a long history of recurring infections, individuals affected with this disorder?
including gastroenteritis, pneumonia, severe staphylococcal 4. How is this disorder transmitted genetically in the
infections, and a liver abscess. He was treated with antibiot- majority of cases?
ics in each case and responded well, albeit slowly. The CBC
was essentially normal, and no morphologic abnormalities Case 2
were detected. His neutrophils were tested repeatedly and A 66-year-old retired male professor is experiencing malaise,
were shown to migrate normally and to respond normally weakness, a fever of 102 F, anorexia, and weight loss. Blood
to chemotactic agents. His neutrophils also phagocytized cultures 8 were negative for pathogens. Ova and parasite
normally; however, they were not able to reduce nitroblue examinations 5 produced negative findings. Results of tuber-
tetrazolium to its insoluble formazan. Minimal conversion culosis testing and fungal serologic testing were negative. The
of dihydrorhodamine to rhodamine was detected by flow CBC revealed a mild normocytic, normochromic anemia,
cytometry. a WBC count of 4.0 109/L, and a platelet count of 130
1. What is the most likely cause of this childs recurring 109/L. The differential count was 38% neutrophil band forms,
infections? 13% neutrophil segmented forms, 20% lymphocytes, 28%
2. Biochemically speaking, what is the specific nature of the monocytes, 0% eosinophils, and 1% basophils. Neutrophils
problem? Continued

408
CHAPTER 28 Nonmalignant Leukocyte Disorders 409

CASE STUDIEScontd
After studying the material in this chapter, the reader should be able to respond to the following case studies:

contained toxic granulation and large vacuoles. Monocytes 2. Where on the blood film should the examination be made
were large and highly vacuolated. The patients erythrocyte and why?
sedimentation rate was 70mm in 1 hour. 3. What is the significance if the suspected cells are found?
1. Based on the differential results, what cells should the 4. What preparation other than a blood film might be helpful
medical laboratory scientist look for on the patients in this situation?
blood film?

N
onmalignant alterations in leukocytes range from receptor. The lamin B receptor is an integral protein in the
relatively rare features (e.g., genetic abnormalities) to inner nuclear membrane,1 and the associated gene is located
findings seen almost on a daily basis, such as leuko- on chromosome band 1q41-43.2 PHA is one of a large number
cyte reactions to a toxic or stressful condition. This chapter of so-called laminopathies ranging from a type of muscular
begins with the rare and finishes with the more common dystrophy to premature aging.3
alterations. Heterozygous PHA is characterized by granulocytes whose
nucleus is segmented only once, taking the form of two very
distinct segments connected by a very thin filament.4 In addi-
GENETIC LEUKOCYTE ABNORMALITIES
tion, what appear to be band forms may be present; however,
Genetic leukocyte abnormalities can be subdivided into those the chromatin shows a highly clumped pattern and stains
that are accompanied by distinct morphologic alterations, and densely. The heterozygous mutation does not affect the func-
functional abnormalities in which morphologic changes are tion of neutrophils.5
often absent. The morphologic abnormalities can be catego- Homozygous PHA is relatively rare and is characterized by
rized into those that affect the nucleus and those that affect the granulocytes with round or oval nuclei (no segmentation) as
cytoplasm. well as the band-form nuclei described earlier. The chromatin
pattern is also very dense and clumped. The homozygous form
Nuclear Abnormalities may be accompanied by impaired cognitive development,
Pelger-Hut Anomaly skeletal abnormalities, and heart defects.5 Leukocyte differen-
Pelger-Hut anomaly (PHA) is an autosomal dominant disor- tial counts performed on blood samples from patients with
der resulting in decreased nuclear segmentation that is most either heterozygous or homozygous PHA may mistakenly iden-
apparent in neutrophils. It is the most common genetic disor- tify the abnormal cells as immature granulocytes if the highly
der of leukocytes and is caused by a mutation of the lamin B clumped, dense chromatin pattern is not noticed (Figure 28-1).

A B
Figure 28-1 Pelger-Hut cells. A, Typical cell found in patients who are heterozygous for Pelger-Hut anomaly (PHA) with two rounded segments connected
by a filament. Notice the dense chromatin pattern. B, Nonsegmented form with dense chromatin that can be seen both in patients who are heterozygous and
in patients who are homozygous for PHA. These cells are sometimes confused with normal neutrophil band forms.
410 PART V Leukocyte Disorders

Figure 28-3 Two neutrophils from a patient with Alder-Reilly anomaly.


Note the dark granules present in both cells. Such granules may also
be seen in eosinophils and basophils. (Courtesy Dennis R. OMalley, MD,
Figure 28-2 Hypersegmented neutrophil such as is seen in patients with US Labs, Irvine, Calif.)
benign hereditary hypersegmentation. These neutrophils should not be con-
fused with the hypersegmented neutrophils seen in megaloblastic anemia. metachromatic granules that may resemble toxic granulation
(From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009, (described later) (Figure 28-3). The granules are composed
Saunders.)
of mucopolysaccharides and are sometimes referred to as
Alder-Reilly bodies or Reilly bodies. In more severe forms, the
Medical laboratory scientists must distinguish inherited granules may also be found in monocytes and lymphocytes.
PHA from an acquired form of nuclear hyposegmentation The basic defect is the incomplete degradation of mucopolysac-
known as pseudoPelger-Hut anomaly (pseudo-PHA) that charides (protein-carbohydrate complexes), and there may be
may be seen in a variety of malignant myeloproliferative a structural abnormality in the myeloperoxidase gene.8 These
neoplasms.6 Two distinctions between inherited PHA and granules are also discussed later in the section on storage
pseudo-PHA are the following: (1) the percentage of affected diseases.
neutrophils is much higher in the inherited form (usually
over 80%), whereas in the acquired form fewer than 50% are Chdiak-Higashi Syndrome
usually affected; and (2) pseudo-PHA alterations are usually Chdiak-Higashi syndrome (CHS) is a rare and fatal autosomal
accompanied by other morphologic indications of malignancy, recessive disease characterized by cells with enlarged lysosomal
such as the presence of blast forms. vesicles. All cells with lysosomal organelles are affected, includ-
ing the melanosomes of melanocytes in the skin, the dense
Hereditary Neutrophil Hypersegmentation granules of platelets, and leukocyte granules. Patients usually
Hereditary neutrophil hypersegmentation designates a group of at have a tendency to bleed, frequent bacterial infections, symp-
least two disorders. The first is a benign autosomal dominant toms of immunodeficiency, variable albinism, and progressive
condition characterized by hypersegmented neutrophils (Figure neurologic dysfunction.9 CHS is associated with a mutation in
28-2) with no clinical signs or symptoms. The major concern the LYST gene that encodes for a type of vesicle trafficking regu-
is to distinguish this condition from the neutrophilic hyperseg- latory protein.10 Patients who do not die of bacterial infection
mentation found in megaloblastic anemia. In the hereditary develop a lymphohistiocytic infiltration into the major organs
form, a large majority of neutrophils are hypersegmented and of the body, resulting in organ failure.9
there is no macrocytic anemia. In megaloblastic anemia, fewer Hematologic findings in CHS include giant lysosomal gran-
than 50% of neutrophils are usually hypersegmented. ules in granulocytes, monocytes, and lymphocytes (Figure
The second disorder is myelokathexis, an inherited form of 28-4). The platelet abnormality usually results in a prolonged
neutropenia characterized by hypersegmented neutrophils and bleeding time, and granule release by thrombin is impaired.11
bone marrow hyperplasia of myeloid cells. There is increased
programmed cell death (apoptosis) of granulocyte precursors.7 May-Hegglin Anomaly
In this case the neutrophil nuclei, in addition to being hyper- May-Hegglin anomaly is one of a group of at least four over
segmented, are pyknotic (to be described later), and the intra- lapping autosomal dominant platelet disorders: May-Hegglin
segmental filaments are longer than normal. anomaly (MHA), Sebastian syndrome (SBS), Fechtner syn-
drome (FS), and Epstein syndrome (EPS). All are caused by
Cytoplasmic Abnormalities mutations of the nonmuscle myosin heavy-chain type IIA gene
Alder-Reilly Anomaly referred to as MYH9.12 MHA, SBS, and FS are characterized by
Alder-Reilly anomaly is transmitted as a recessive trait and large basophilic inclusions in leukocytes, thrombocytopenia,
is characterized by granulocytes with large, darkly staining and giant platelets (Figure 28-5). FS and EPS are also associated
CHAPTER 28 Nonmalignant Leukocyte Disorders 411

A B

C
Figure 28-4 Three cells from a patient with Chdiak-Higashi syndrome. A, Neutrophil with large dark lysosomal granules. B, Monocyte with large azure
granules. C, Lymphocyte with one large azure granule.

chapter); however, there are striking ultrastructural differences.


Dhle bodies are made up of lamellar rows of rough endoplas-
mic reticulum, whereas the MHA inclusions consist of ran-
domly placed rods in an amorphous background. In addition,
Dhle bodies are found only in neutrophils, whereas MHA
inclusions are seen in neutrophils, eosinophils, basophils, and
monocytes.14 MHA inclusions may stain a very pale color with
Wright stain and may be missed in monocytes whose cyto-
plasm is also blue-grey.
Clinically, the majority of individuals with MHA are asymp-
tomatic; however, a few have had bleeding episodes, possibly
related to the thrombocytopenia.

Functional Abnormalities
The majority of genetic functional leukocyte disorders, with the
Figure 28-5 A neutrophil and a giant platelet from a patient with May- exception of some of the storage disorders, are not character-
Hegglin anomaly. Note the large, elongated, bluish inclusion in the neutrophil ized by specific morphologic alterations in leukocytes.
cytoplasm.
Chronic Granulomatous Disease
with nephritis and deafness. Patients with MHA and SBS gener- Chronic granulomatous disease (CGD) is a disorder caused by
ally do not have renal disease or deafness; however, there have the inability of phagocytes to produce superoxide and reactive
been a few reports of renal disease in individuals with MHA.13 oxygen species. The basic defect is one or more mutations in
The leukocyte inclusions in MHA have been described as any of four genes responsible for proteins that make up a
Dhle bodylike (Dhle bodies are discussed later in this complex known as NADPH oxidase (NADPH is the reduced
412 PART V Leukocyte Disorders

A B
Figure 28-6 Two nitroblue tetrazolium (NBT) preparations. A, Neutrophils from a normal control that have reduced the NBT to a dark formazan.
B, Neutrophils from a patient with chronic granulomatous disease. (Courtesy Valerie Evans, University of Arizona Medical Center, Tucson, Ariz.)

form of nicotinamide adenine dinucleotide phosphate). The retardation, growth defects, and/or deficiency in H blood
majority of cases (approximately 60% to 65%) are X-linked group antigen.17 In this case, the basic defect is faulty ligands
recessive and 35% to 40% are autosomal recessive, depending for selectin adhesive molecules.
on which gene has been mutated. The X-linked forms tend to Patients with LAD-III present with Glanzmann
carry a higher mortality rate.15 thrombasthenialike bleeding problems (see Chapter 44) as
CGD results in a predisposition to bacterial and fungal well as LAD-I symptoms despite normal integrin expression. A
infections and the formation of granulomas that can obstruct recent report indicates that the basic defect is a mutation in
hollow organs such as the stomach, intestines, and urinary KINDLIN3, a gene responsible for a protein that binds to the
tract. Catalase-negative bacteria are rarely involved in CGD 2 integrin subunit.18
infections due to the fact that they generate their own hydrogen
peroxide within the lysosome and can thus be killed by the Miscellaneous Granulocyte Disorders
neutrophil phagocytes. Miscellaneous granulocyte disorders include several relatively
In the nitroblue tetrazolium reduction test, normal neutro- rare diseases.
phils, when stimulated, reduce the yellow water-soluble nitro- Hyperimmunoglobulinemia E, also known as Job syndrome, is
blue tetrazolium to a dark blue insoluble formazan. CGD characterized by elevated levels of immunoglobulin E, skeletal
neutrophils cannot perform this reduction (Figure 28-6). Flow abnormalities, and recurrent severe bacterial infections.19
cytometry uses a fluorescent probe, such as dihydrorhodamine- Neutrophils in these patients have poor directional motility.
123, to measure intracellular production of reactive oxygen Evidence points to dysregulation in cytokine production by T
species.15 cells, resulting in imbalances between interferon- and trans-
forming growth factor in relation to interleukin-4 (IL-4).20
Leukocyte Adhesion Disorders Lazy leukocyte syndrome is extremely rare and is characterized
Leukocyte adhesion disorders (LADs) result in the inability of by neutropenia and poor neutrophil response to chemotactic
neutrophils and monocytes to adhere to endothelial cells and agents.21 Abnormalities in neutrophil cytoplasmic actin and
to transmigrate from the blood to the tissues. The consequence myosin microfilaments are responsible for the dsyfunction.22
of this inability is increased and potentially lethal bacterial Myeloperoxidase (MPO) deficiency is probably the most
infections. The basic defect is a mutation in the genes respon- common neutrophil abnormality; however, symptoms are
sible for the formation of cell adhesion molecules, including relatively mild in most patients. The discovery of MPO defi-
integrins and selectins (see Chapter 12). LADs have been sub- ciency is credited to the automated hematology analyzers that
divided into three subcategories. LAD-I is caused by a mutation use MPO to identify cells. The MPO gene is located on chromo-
in the gene(s) responsible for 2 integrin subunits (CD11/ some 17, and up to 10 mutations have been described.23
CD18), resulting in either decreased or defective 2 integrins, Diabetes mellitus can be either inherited or acquired. It has
which are necessary for adhesion to endothelial cells, recogni- been associated with poor neutrophil function, especially
tion of bacteria, and outside-in signaling.16 In addition to expe- when glucose levels are very high.24 The most consistent abnor-
riencing recurrent infections, patients with LAD-I frequently mality is abnormal oxidative burst activity.25
have neutrophilia.
LAD-II is considerably rarer than LAD-I and presents in a Monocyte/Macrophage Lysosomal Storage Diseases
similar manner (recurrent infection, neutrophilia, and impaired Monocyte/macrophage lysosomal storage diseases can be subdi
pus formation); however, the leukocytes have normal 2 inte- vided into mucopolysaccharide (or glycosaminoglycan [GAG])
grins. In addition, patients with LAD-II may have mental storage diseases and lipid storage diseases (Table 28-1). As a
CHAPTER 28 Nonmalignant Leukocyte Disorders 413

TABLE 28-1 Variants of Monocyte/Macrophage Lysosomal Storage Disorders


Type Name Deficient Enzyme Substance Stored
Mucopolysaccharidosis
MPS Isevere Hurler syndrome -l-iduronidase Dermatan sulfate, heparan sulfate
MPS Iattenuated Scheie syndrome -l-iduronidase Dermatan sulfate, heparan sulfate
MPS II severe Hunter syndrome Iduronate sulfatase Dermatan sulfate, heparan sulfate
MPS III Sanfilippo syndrome type A Heparan N-sulfatase Heparan sulfate
Sanfilippo syndrome type B -N-acetylglucosaminidase Heparan sulfate
Sanfilippo syndrome type C Acetylcoenzyme A:-glucosaminide Heparan sulfate
N-acetyltransferase
MPS IV Morquio syndrome type A Galactose-6-sulfatase Keratan sulfate, chondroitin-6-sulfate
Morquio syndrome type B -Galactosidase Keratan sulfate
Lipid storage diseases
Gaucher disease -glucocerebrosidase Glucocerebroside
Niemann-Pick disease Sphingomyelinase Sphingomyelin
Fabry disease -Galactosidase Ceramide trihexoside
Tay-Sachs disease, Sandhoff disease Hexosaminidase A GM2 ganglioside

group, they represent inherited enzyme deficiencies or defects


that result in flawed degradation of phagocytized material and
buildup of the partially digested material within the phagocyte.
All cells containing lysosomes can be affected, including T
lymphocytes.26
The mucopolysaccharidoses (MPSs) are a family of inherited
disorders of GAG degradation. Each MPS is caused by deficient
activity of an enzyme necessary for the degradation of derma-
tan sulfate, heparan sulfate, keratan sulfate, and/or chondroi-
tin sulfate. The partially degraded GAG builds up in the
lysosomes and eventually results in physical abnormality and
sometimes mental retardation. The MPSs have been subdi-
vided according to which enzyme is defective, which GAG is
being stored, and whether the symptoms are severe or attenu-
ated (see Table 28-1).27
The peripheral blood of a patient with MPS may appear
relatively normal; however, metachromatic Reilly bodies may
be seen in neutrophils, monocytes, and lymphocytes (Figure Figure 28-7 Lymphocyte on the blood film for a patient with a mucopoly-
28-7). Bone marrow may reveal macrophages with large saccharide storage disorder known as Hurler disease. Notice the dark
amounts of metachromatic material. Diagnosis relies on assays granules that appear to be in vacuoles within the cytoplasm.
for the specific enzymes involved. Treatment has consisted of
enzyme replacement therapy or hematopoietic stem cell these patients. Bone marrow contains distinctive macrophages
transplantation.27 called Gaucher cells, occurring individually or in clusters, that
Lipid storage diseases include a variety of disorders in which have abundant fibrillar blue-grey cytoplasm with a striated or
lipid catabolism is defective (Figure 28-8). Two of these disor- wrinkled appearance (sometimes described as onion skinlike)
ders are characterized by macrophages with distinctive mor- (Figure 28-9).
phology and are discussed here. Treatment of Gaucher disease includes the use of recombi-
Gaucher disease is the most common of the lysosomal lipid nant glucocerebrosidase, which is quite expensive.29 Another
storage diseases. It is an autosomal recessive disorder caused approach is the use of pharmacologic chaperones.30
by a defect in the catabolic enzyme -glucocerebrosidase. This Pseudo-Gaucher cells that look remarkably similar to
results in accumulation of glucocerebroside in macrophages Gaucher cells under the light microscope can be seen in a
throughout the body, including osteoclasts in bone and variety of conditions in which cell turnover is increased. For
microglia in the brain. Gaucher disease has been subdivided example, they have been reported in thalassemia,31 chronic
into three types based on clinical signs and symptoms (Table myelogenous leukemia,32 and acute lymphoblastic leukemia.33
28-2).28 Hematologic features may include anemia and throm- Pseudo-Gaucher cells reflect an overwhelming of the glu
bocytopenia reflecting the hypersplenism that is common in cocerebrosidase enzyme by the excessive cell turnover rather
414 PART V Leukocyte Disorders


cer glc gal galNac gal

Nana
GM1-ganglioside

GM1 b-galactosidase
GM1-GANGLIOSIDOSIS
cer glc gal gal galNac
globoside

cer glc gal galNac Hexosaminidases A and B


SANDHOFFS DISEASE
Nana
GM2-ganglioside

Hexosaminidase A
TAY-SACHS DISEASE cer glc gal gal
SANDHOFFs DISEASE ceramide trihexoside

a-Galactosidase
cer glc gal FABRYS DISEASE
Nana
GM3-ganglioside
cer glc ga
ceramide lactoside

cer gal SO4


sulfatide
cer glc
Arylsulfatase A
glucocerebroside
METACHROMATIC
LEUKODYSTROPHY

Glucocerebrosidase
cer gal GAUCHERS DISEASE
galactocerebroside cer p choline
sphingomyelin

Galactocerebrosidase
b-Galactosidase
KRABBES DISEASE cer Sphingomyelinase
ceramide NIEMANN-PICK DISEASE

Ceramidase
FARBERS DISEASE

sphingosine and fatty acid

Figure 28-8 Pathways and diseases of sphingolipid metabolism. (From Orkin SH, Fisher DE, Look AT, etal: Nathan and Oskis hematology of infancy and
childhood, ed 7, Philadelphia, 2009, Saunders.)

TABLE 28-2 Clinical Subtypes of Gaucher Disease


Clinical Features Type I: Nonneuropathic Type II: Acute Neuropathic Type III: Subacute Neuropathic
Clinical onset Childhood/adulthood Infancy Childhood
Hepatosplenomegaly + + +
Skeletal abnormality + +
Neurodegeneration +++ ++
Death Variable By 2 years 2nd-4th decade
Ethnic predilection Ashkenazi Jews Panethnic Swedes

NOTE: Absence and severity of features are indicated by to +++.


CHAPTER 28 Nonmalignant Leukocyte Disorders 415

Figure 28-9 Characteristic macrophages with cytoplasmic striations


found in the bone marrow of a patient with Gaucher disease. (From Carr JH,
Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009, Saunders.)
Figure 28-10 Niemann-Pick cell with eccentric nucleus and bubblelike
pattern of storage deposit in the cytoplasm. (From Carr JH, Rodak BF: Clinical
than a true decrease in the enzyme. Electron microscopy can hematology atlas, ed 3, St Louis, 2009, Saunders.)
distinguish between the two cells, because pseudo-Gaucher
cells do not contain the tubular inclusions described in T-lymphocyte abnormality is best represented by a condition
Gaucher cells. known as DiGeorge syndrome. This syndrome is characterized
Niemann-Pick disease is a group of disorders that are subclas- by the absence or underdevelopment of the thymus and hence
sified into two categories, those with a primary deficiency in markedly decreased numbers of T lymphocytes. It is associated
acid sphingomyelinase (ASM) and those with defective NPC1 with a microdeletion in chromosome band 22q11.2, which is
and NPC2 genes that regulate intracellular processing and the most common chromosome deletion and occurs in approx-
transport of low-density lipoproteinderived cholesterol.34 imately 1 in 3500 births. Individuals with this deletion have a
ASM deficiency occurs in a broad clinical spectrum ranging broad range of abnormalities, including defective parathyroid
from an infantile neurologic form that results in death by glands, cardiac abnormalities, abnormal facial development,
3 years of age to a nonneurologic form compatible with sur- and neurologic disorders. Fewer than 1% of patients with this
vival into adulthood, with numerous intermediate forms in deletion are athymic, a condition sometimes referred to as
between.35 ASM is a lysosomal enzyme whose substrate is complete DiGeorge syndrome.39 Many of these patients are treated
sphingomyelin, which accumulates in the lysosome if ASM is with thymus transplantation, with an approximately 75%
defective. Over 100 mutations have been reported in the gene survival rate.40
for ASM. The bone marrow of individuals with Niemann-Pick B-lymphocyte abnormality is represented by sex-linked agam-
disease contains numerous macrophages with a foamy cyto- maglobulinemia (XLA), which is caused most frequently by a
plasm packed with lipid-filled lysosomes that appear as mutation in the gene encoding Bruton tyrosine kinase (Btk).
vacuoles after staining (Figure 28-10). Although there is no Such mutations result in a block in B-lymphocyte develop-
satisfactory treatment at this time, phase 1 studies of enzyme ment.41 A large number of different mutations in the Btk gene
replacement are being carried out, and gene therapy is being have been reported; however, other mutations in the preB cell
evaluated in mice.35 receptor complex proteins have also been reported in patients
Discovery of the NPC1 gene36 in 1997 and a second (NPC2) with presumed XLA, such as mutations in the heavy chain.42
gene37 in 2000 began a new wave of research and study on Combined B-lymphocyte/T-lymphocyte abnormalities include
Niemann-Pick disease. The functions of the encoded proteins severe combined immunodeficiency (SCID) and Wiskott-
are unknown. When NPC1 protein is defective, there is Aldrich syndrome. SCID can be divided into two types: adenos-
an accumulation of sphingosine, glycosphingolipids, sphingo ine deaminase deficiency and X-linked SCID. Both result
myelin, and cholesterol. Macrophages as described earlier can in depletions of T, B, and natural killer (NK) lymphocytes.
be found in most organs. One hypothesis is that the accumula- Adenosine deaminase deficiency results in excess amounts of
tion of sphingosine causes a decrease in lysosomal calcium, its natural substrates (adenosine and 2-deoxyadenosine),
which, in turn, leads to defective transport and accumulation which cause lymphocyte depletion through a variety of
of cholesterol.38 mechanisms.43
X-linked SCID is the more common of the two and is caused
B- and T-Lymphocyte Abnormalities by a mutation in the gene encoding the IL-2 receptor chain,
B- and T-lymphocyte abnormalities are genetic disorders that which is shared by several interleukins. The mutation results
generally result in the decreased production of B cells, T cells, in T lymphopenia, B cells that are dysfunctional, and a lack of
or both. NK cells.44
416 PART V Leukocyte Disorders

Wiskott-Aldrich syndrome is also X-linked and is caused by An absolute decrease in neutrophils below 2.0 109/L is
a mutation in a gene that encodes a protein called WASp. The referred to as neutropenia. Neutropenia can be genetic or
mutation results in low levels or absence of the protein, and acquired. Genetic neutropenias (often referred to as congenital
affected individuals have immunodeficiency, eczema, and neutropenias) are a relatively rare group of disorders associated
thrombocytopenia. Absence of WASp affects migration, adhe- with decreased marrow production of neutrophils, often due
sion, and activation of a variety of leukocytes, including T cells, to a mutation in ELA2, the gene coding for neutrophil elastase,
B cells, and NK cells.45 and less frequently due to mutations in hematopoietic regula-
tory genes.46,47
Acquired neutropenia can result from decreased production
REACTIVE OR STRESS-RELATED
caused by marrow aplasia, acute leukemia, or a megaloblastic
LEUKOCYTE ABNORMALITIES
process. The presence of autoimmune antibodies against neu-
Quantitative Abnormalities trophils leads to neutropenia. Neutropenia can also be due to
Neutrophils overwhelming infection and inflammation, which are often
An absolute increase in neutrophils above 8.7 109/L is seen in immunocompromised patients and in the elderly.
referred to as neutrophilia and can be caused by a shift in neu- Table 28-3 lists causes of neutropenia.
trophils from the marginated pool to the circulating pool, such
as is seen after strenuous exercise, during labor, in tachycardia, Eosinophils
and with epinephrine or cortisone therapy. It can also be Eosinophilia is defined as an absolute eosinophil count above
caused by pathologic conditions that result in a shift from the 0.4 109/L.
bone marrow storage pool to the peripheral blood and Increased eosinophils are seen in patients with parasite
increased production of neutrophils. The latter is almost always infestations, especially those due to tissue-invading parasites.
accompanied by a shift to the left (presence of immature neu- Eosinophils are also increased in allergic disorders such as
trophils). Table 28-3 lists the major types of disorders that asthma48 and dermatitis.49
result in neutrophilia. Certain tumors cause an increase in eosinophils both in
The term leukemoid reaction refers to a leukocyte count above the blood and around the tumor. Eosinophils are able to
50 109/L with neutrophilia and a marked left shift. Leuke- penetrate solid tumors, which allows tumoricidal cytokines to
moid reactions can be confused with chronic myelogenous bring about tumor necrosis.50 Certain autoimmune disorders
leukemia. There are several distinguishing features between the such as lupus erythematosus have also been associated with
two, and these are listed in Box 28-1. eosinophilia.51
The term leukoerythroblastic reaction refers to the presence of If an individual is found to have eosinophilia without any
immature neutrophils and nucleated red blood cells in the discernible cause such as those listed above, and if the eosino-
same sample. Leukoerythroblastic reactions are often accom- philia lasts more than 6 consecutive months, the diagnosis of
panied by neutrophilia, but not necessarily. Leukoerythroblas- hypereosinophilic syndrome, or HES, is used. HES is rare and is
tic reactions point to the probability of a space-occupying associated with damage to organs such as the skin, heart, and
lesion in the bone marrow. A space-occupying lesion can be a lungs as well as central and peripheral nervous systems. Con-
metastatic tumor, fibrosis, lymphoma, leukemia, or simply a sequently, once a diagnosis of HES is made, corticosteroids and
marked increase in one of the normal marrow cells (e.g., the
erythroid hyperplasia seen in hemolytic anemia).
BOX 28-1 Distinguishing Between Chronic
Myelogenous Leukemia and Leukemoid Reaction
TABLE 28-3 Causes of Neutrophilia and Neutropenia
Chronic Myelogenous Leukemia
Neutrophilia Neutropenia
Increases in all granulocytes including eosinophils and basophils and
Physiologic (e.g., strenuous Overwhelming infection their immature forms
exercise) Hemodialysis Dyspoietic morphology such as mixed granulation or pseudo
Acute infections Physical agents/drugs (e.g., Pelger-Hut
Inflammation (e.g., burns, after chemotherapy, radiation, Involvement of platelets including giant, hypogranular forms
surgery, myocardial infarction) chloramphenicol, any agent Leukocyte alkaline phosphatase score is markedly decreased
Intoxication (e.g., uremia, diabetic causing aplasia)
acidosis, poisoning, insect Decreased or ineffective production Leukemoid Reaction
envenomation) (e.g., megaloblastic anemia) Increases in neutrophils and their immature forms; possible increased
Acute hemorrhage or hemolysis Splenic sequestration monocytes but eosinophils and basophils are frequently absent
Response to fast-growing tumors Autoimmune disorders No dyspoietic morphology; however, reactive morphology is usually
Malignant myeloproliferative Debilitated states (e.g., alcoholism) present
disorders Hereditary disorders (e.g., cyclic No abnormal platelet morphology
Certain medications (e.g., steroids) neutropenia) Leukocyte alkaline phosphatase score is markedly increased
CHAPTER 28 Nonmalignant Leukocyte Disorders 417

TABLE 28-4 Causes of Eosinophilia and Eosinopenia TABLE 28-5 Causes of Monocytosis and
Monocytopenia
Eosinophilia Eosinopenia
Monocytosis Monocytopenia
Allergic reactions (e.g., asthma, hay Infections or inflammation in
fever, drug sensitivity) which neutrophilia occurs Bacterial infections (e.g., Overwhelming infections in
Parasitic infestations (especially with Thymoma and tuberculosis, subacute bacterial immunocompromised patients
tissue-invading parasites) hypogammaglobulinemia endocarditis, syphilis) Hemodialysis
Hypersensitivity reactions (e.g., Rare inherited forms Recovery from acute infection Epstein-Barr virus infection
Loeffler syndrome) Autoimmune disorders with Protozoal and rickettsial infections Steroid therapy
Skin diseases (e.g., dermatitis antieosinophil antibodies (e.g., malaria, typhus)
herpetiformis) Unknown causes Certain carcinomas
Tropical eosinophilia Granulomatous diseases
Malignant myeloproliferative disorders Collagen vascular diseases (e.g.,
Certain infections (e.g., scarlet fever) lupus erythematosus)
Malignancies of monocyte cell line

monoclonal antibodies against IL-5 are administered to lower


the eosinophil count.52 Table 28-4 lists the major nonmalig- Certain protozoal and rickettsial infections such as malaria
nant causes of eosinophilia and eosinopenia. and Rocky Mountain spotted fever may be associated with
Eosinopenia is defined as an absolute eosinophil count of monocytosis. Noninfectious diseases associated with monocy-
less than 0.09 109/L. The most common cause of decreased tosis include collagen vascular diseases such as lupus erythe-
eosinophils is infection or inflammation that is accompanied matosus, and malignancies of the ovary, stomach, and breast
by neutrophilia. Eosinophils move into the tissues under these are frequently associated with monocytosis.56
circumstances, and marrow release of eosinophils may be Monocytopenia, defined as an absolute monocyte count of
inhibited. More rare causes of eosinopenia include autoim- less than 0.4 109/L, can be seen following steroid therapy57
mune disorders that result in the formation of antieosinophil as well as during hemodialysis.58 Viral infections, especially
antibodies, hypogammaglobulinemia in the presence of those due to the Epstein-Barr virus (EBV), can cause monocy-
thymoma, certain drug allergies, and urticaria.53 topenia (Table 28-5).59

Basophils Lymphocytes
Basophilia is defined as an absolute basophil count greater The definition of lymphocytosis varies with the age of the indi-
than 0.15 109/L. The most common cause of basophilia vidual. Children older than 2 weeks and younger than 8 to 10
is the presence of a malignant myeloproliferative neoplasm years normally have higher lymphocyte counts than adults.
such as chronic myelogenous leukemia, which is covered in Lymphocytosis in young children is defined as an absolute
Chapter 34. Nonmalignant causes of basophilia are relatively lymphocyte count greater than 10.0 109/L, whereas in adults,
rare and include hypothyroidism, ulcerative colitis, some it is defined as a count greater than 4.8 109/L. As can be seen
bee stings, and some types of nephrosis. Because the normal from the tables inside the front cover of this book, newborn
number of basophils is so low, basopenia is difficult to infants have lymphocyte counts very similar to those of adults.
establish. Basopenia has been reported, however, in chronic Lymphocytoses can be subdivided into those with and those
urticaria.54 without reactive morphologic alterations, as are described
later. See Table 28-6 for a listing of disorders that result in
Monocytes lymphocytosis. Infectious lymphocytosis is a childhood disease
Monocytosis is defined as an absolute monocyte count greater characterized by large numbers of normal-looking small
than 1.1 109/L. Because monocytes are not stored in the bone lymphocytes. If lymphoid malignancies are accompanied
marrow, relative monocytosis is frequently the first sign of by morphologic alterations, the alterations are not reactive
recovery from acute overwhelming infection or severe neutro- but dyspoietic and are often clonal in nature (i.e., all cells
penia (agranulocytosis). Monocytosis is also seen during look alike).
certain bacterial infections, especially tuberculosis, subacute The definition of lymphocytopenia is also age dependent.
bacterial endocarditis, syphilis, and brucellosis. In addition to Lymphocytopenia in young children is defined as an absolute
monocytosis, subacute bacterial endocarditis is often character- lymphocyte count below 2.0 109/L, whereas in adults, it is
ized by the presence of circulating macrophages that can be defined as a count below 1.0 109/L. The most common cause
seen in the edges of blood films due to their large size. Circulat- of lymphopenia is treatment with steroids. Another relatively
ing macrophages can be found weeks before the blood culture common acquired cause is infection with human immuno
is positive for an infectious agent.55 Some laboratories make deficiency virus (HIV), which targets CD4 lymphocytes. Less
nucleated cell concentrate films (buffy coat preparations) to common causes include the genetic lymphocyte abnormalities
increase the chance of finding the macrophages. described earlier in this chapter.
418 PART V Leukocyte Disorders

Qualitative (Morphologic) Abnormalities (Figure 28-11). Eosinophils may also become hypersegmented
Granulocytes (three or more nuclear segments) when reacting to a toxic
Granulocyte reaction to infection or inflammation includes the situation.
presence of immature forms in the circulation, referred to as a Pyknotic nuclei in neutrophils generally indicate imminent
left shift, as well as morphologic alterations affecting both cell death. In a pyknotic nucleus, nuclear water has been lost
nucleus and cytoplasm. Depending on the severity of the and the chromatin becomes very dense and dark; however, fila-
insult, the left shift in such situations can range from mild (an ments can still be seen between segements.60 Pyknotic nuclei
increase in band forms and an occasional metamyelocyte), to should not be confused with necrotic nuclei found in dead
moderate (presence of metamyelocytes, myelocytes, and an cells. Necrotic nuclei are rounded fragments of nucleus with
occasional promyelocyte), to marked (myelocytes, promyelo- no filaments and no chromatin pattern (Figure 28-12).
cytes, and an occasional blast form). Cytoplasmic abnormalities in granulocytes caused by toxic
Nuclear abnormalities caused by reaction to infection, inflam- conditions include Dhle bodies, toxic granulation, vacuola-
mation, or stress include nuclear hypersegmentation and pyk- tion, degranulation, and swelling.
nosis. Hypersegmentation is often seen in chronic infections Dhle bodies are cytoplasmic inclusions consisting of ribo-
and may reflect borderline folate deficiency caused by long- somal ribonucleic acid (RNA) arranged in parallel rows.61
term hyperproliferation of granulocytes in response to the Dhle bodies are found in band and segmented neutrophils
infection or inflammation. Toxic hypersegmentation is differ- (Figure 28-13). With Wright staining, they appear as pale blue
entiated from that seen in megaloblastic anemia because it
is not accompanied by the presence of macrocytic red cells

TABLE 28-6 Causes of Lymphocytosis and


Lymphocytopenia
Lymphocytosis Lymphocytopenia
Without Morphologic Steroid therapy
Alterations Strenuous exercise
Bordetella pertussis infection Morphine administration
Acute infectious lymphocytosis Human immunodeficiency
virus infection
With Morphologic Alterations Genetic abnormalities
Infectious mononucleosis
Infectious hepatitis
Cytomegalovirus infection
Viral influenza Figure 28-11 Neutrophil from a patient with septicemia showing toxic
Lymphoid malignancies hypersegmentation (at least seven segments). Other evidence of toxicity is
the presence of cytoplasmic vacuoles.

A B
Figure 28-12 A, Upper cell is a neutrophil whose nucleus is dehydrated, which makes it very dark and dense. Note that there is still a filament between
the segments. This is referred to as a pyknotic cell. The cell is also highly vacuolated. B, Former neutrophil that has died. Note that the nucleus has disinte-
grated into numerous rounded spheres of DNA with no filaments seen. This is referred to as a necrotic or necrobiotic cell. (B from Carr JH, Rodak BF: Clinical
hematology atlas, ed 3, St. Louis, 2009, Saunders.)
CHAPTER 28 Nonmalignant Leukocyte Disorders 419

round or elongated bodies between 1 and 5m in diameter. MPS (see earlier in chapter). Unlike the dark granules seen in
They are usually located in close apposition to cellular mem- the other two situations, true toxic granules tend to cluster
branes. Any time delay in preparing the blood film from the within the neutrophil cytoplasm, and not all neutrophils are
ethylenediaminetetraacetic acid (EDTA) tube can affect their equally affected (Figure 28-14).
appearance so that they may look more gray than blue and in Vacuolation in granulocytes generally reflects phagocytosis,
some cases may not be visible. Dhle bodies are relatively either of self (autophagocytosis) or of extracellular material.
nonspecific. Their presence has been associated with a wide Autophagocytosis can be caused by drugs such as sulfonamides
range of conditions, including bacterial infections, sepsis,61 and and chloroquine,59 by prolonged storage in EDTA, by auto
normal pregnancy.62 antibodies,67 by acute alcoholism,68 and by exposure to high
Toxic granulation reflects an increase in mucosubstance doses of radiation.69 Autophagocytic vacuoles tend to be small
within primary granules.63 There is a positive correlation (approximately 2m) and distributed throughout the cyto-
between levels of C-reactive protein and the percentage of neu- plasm. Phagocytic vacuoles are often seen in septic processes
trophils with toxic granulation64; therefore, toxic granulation is caused by either bacteria or fungi. Phagocytic vacuoles are large
considered an important marker of inflammation.65 Toxic gran- (up to 6m) and are frequently outlined by visible toxic gran-
ulation may be induced by increases in granulocyte colony- ules (Figure 28-15). It is important to distinguish between true
stimulating factor.66 Toxic granules are larger than specific vacuolation and that caused by excessive storage in EDTA. If
granules and stain blue-black with Romanowsky stains. It is
important to distinguish between true toxic granulation, poor
staining, and the metachromatic granules found in inherited

Figure 28-14 Toxic granulation. Note that one neutrophil contains toxic
granulation and the other does not. Also note that the toxic granules are
clustered in some areas of the cytoplasm. Both of these findings help in
Figure 28-13 Neutrophil containing a bluish cytoplasmic inclusion known distinguishing toxic granulation from poor staining or from the dark granules
as a Dhle body. seen in Alder-Reilly anomaly.

A B
Figure 28-15 Cytoplasmic vacuoles. A, Band form neutrophil with autophagocytic vacuoles. Note their small size. B, Neutrophil with phagocytic vacuoles.
Note the much bigger size. Other evidence of toxicity in this cell is the pyknotic nucleus and the fact that the cytoplasm appears degranulated.
420 PART V Leukocyte Disorders

A
B

Figure 28-16 A, Anaplasma phagocytophilum in a neutrophil. B, Ehrlichia


chaffeensis in a monocytic cell. (Courtesy J. Stephen Dumler, MD, Division
of Medical Microbiology, The Johns Hopkins Medical Institutions, Baltimore,
Md.)

Figure 28-18 Neutrophil anisocytosis. The neutrophil to the left is larger


than the other neutrophil. This is often caused by cytoplasmic swelling.

Figure 28-17 Partially degranulated eosinophil. This cell was found on


the blood film for a patient with trichinosis.

Figure 28-19 Immature monocyte. This cell was found on the blood film
the blood film was made more than 2 hours after venipuncture, from a police officer who had been shot and had sepsis. The patient had an
artifactual vacuolation should be suspected. absolute monocytosis with numerous reactive forms.
When phagocytic vacuoles are seen, a careful examination
should be made for bacteria or yeast within the vacuoles. Cases
of ehrlichiosis and anaplasmosis have been increasing in the secondary granules are emptied into phagosomes, and second-
United States over the past decade.70-72 Ehrlichia and Anaplasma ary granules are also secreted into the extracellular space.74
are small obligate intracellular bacteria transmitted by ticks to Degranulation is often accompanied by disruption of cellular
humans and other vertebrate hosts. These organisms grow as membranes during the process of making the blood film.
a cluster (morula) in neutrophils (A. phagocytophilum and E. Cytoplasmic swelling may be caused by actual osmotic
ewingii) (Figure 28-16, A) and in monocytes (E. chaffeensis) swelling of the cytoplasm or by increased adhesion to the glass
(Figure 28-16, B). Leukopenia, thrombocytopenia, and ele- slide by stimulated neutrophils. Regardless of cause, the result
vated liver enzymes are common laboratory findings, and is a variation in neutrophil size, or neutrophil anisocytosis
anemia may develop in about half the cases of human mono- (Figure 28-18).
cytic ehrlichiosis after 2 weeks.70,73 Intracellular aggregates in
neutrophils or monocytes may occasionally be detected in the Monocytes
first week of infection on a Wright-Giemsastained peripheral Occasional immature monocytes may be seen in the peripheral
blood film or a buffy coat preparation, but immunofluorescent blood in response to infection or inflammation, but this is not
antibody titers or polymerase chain reaction testing is needed as common as neutrophilic left shifts (Figure 28-19).
for confirmation.70 Early diagnosis is essential, because antibi- Nuclear alterations in activated monocytes are not common.
otic treatment with doxycycline is very effective and can prevent Nuclear contortion is the only nuclear alteration that is
serious complications. described here. In these cases, the monocyte nucleus becomes
Degranulation is a common finding in activated neutro- quite thin and bandlike in areas and may appear to be seg-
phils and eosinophils (Figure 28-17). Both primary and menting (Figure 28-20).
CHAPTER 28 Nonmalignant Leukocyte Disorders 421

Figure 28-20 Reactive monocyte with contorted nucleus. Other evidence Figure 28-22 Reactive lymphocyte, probably a B cell, showing evidence
of toxicity is the several vacuoles in the cytoplasm. of blastogenesis. Note the loose chromatin pattern and the two (possibly
three) nucleoli within the nucleus.

normal way. The terms variant and reactive are often used
interchangeably.
Nuclear alterations in reactive lymphocytes depend on the
stage of blastogenesis and whether the cell is a T lymphocyte
or a B lymphocyte. When a small resting lymphocyte is stimu-
lated by the presence of antigen, it begins to enlarge, becoming
a medium-sized lymphocyte and then a large lymphocyte.
Meanwhile the chromatin pattern becomes less clumped and
more homogeneous until nucleoli appear and the cell is rec-
ognized as a blast. A general rule of thumb when performing
differential counts and encountering what looks like a blast
form is that if all other findings are normal, the blast form is
most likely a reactive lymphocyte undergoing blastogenesis
and not anything more sinister (Figure 28-22).
Figure 28-21 Monocyte showing evidence of transformation into a mac- If the cell is a B lymphocyte, the blast form could be called
rophage. Note the increased cytoplasmic volume, the large number of vacu- an immunoblast. The blast divides to form a memory cell for
oles, and the irregular cytoplasmic borders. that particular antigen and a proplasmacyte that then matures
into a plasma cell or a plasmacytoid lymphocyte with its highly
clumped nuclear chromatin. (See Figure 12-19.)
The cause of nuclear contortions in monocytes is unknown; If the cell is a T lymphocyte, the blast divides to form a
however, they are frequently seen on blood films in which toxic memory cell for the particular antigen and an effector T cella
neutrophil hypersegmentation is also occurring. Hence, they wide-bodied cell that is sometimes referred to as an infectious
may also reflect borderline folate deficiency. mononucleosis cell or viral lymphocyte because it is a cell charac-
Cytoplasmic alterations in toxic monocytes are those changes teristic of infectious mononucleosis. The nucleus of the effector
associated with transformation into macrophages. These T cell is somewhat fragile and has a tendency to adhere to the
include increases in cytoplasmic volume and spreading ability, inner plasma membrane, and this effect becomes quite exag-
increased numbers of cytoplasmic granules, and evidence gerated when the sample is held in EDTA for a prolonged
of phagocytic activity (cytoplasmic vacuolation, intracellular period (Figure 28-23).
debris, and highly irregular cytoplasmic borders) (Figure 28-21). Cytoplasmic alterations in reactive lymphocytes are mainly
increases in cytoplasmic RNA. If the cell is an effector B cell,
Lymphocytes which is a synthesizer of protein, there is considerable RNA in
Over the past century, reactive alterations in lymphocytes the cytoplasm and it therefore appears very blue with
have been described by several confusing terms, including Romanowsky staining. If the cell is an effector T cell, the RNA
variant lymphocytes, reactive lymphocytes, effector lymphocytes, appears to be localized in patches of blue along the periphery
and atypical lymphocytes. The last designation is the least of the cell as well as radiating out from the nucleus. Because
suitable, because it implies that the lymphocytes are abnor- of this, the reactive T cell has been said to look like a flared
mal when, in fact, they are reacting to antigen in a very skirt or a fried egg. Another cytoplasmic feature seen in
422 PART V Leukocyte Disorders

A B
Figure 28-23 Reactive lymphocytes of the T-cell variety found on the blood films for patients with infectious mononucleosis. A, T-reactive lymphocyte on
a blood film that was made within 2 hours of venipuncture. Note the flared-skirt appearance of the cytoplasm and the apparent bonding of the nucleus to the
cytoplasmic membrane in one spot. B, Two reactive lymphocytes on a blood film that was made over 4 hours after venipuncture. Note the exaggerated nuclear
fragility.

pharyngitis (sore throat), fever, and lymphadenopathy.76


Fatigue, malaise, chills and fever, loss of appetite, and nausea
are frequently cited. Splenomegaly is common but often
only detected by ultrasound examination. Although there
may be some elevation of liver function tests, hepatomegaly is
rare. The majority of patients recover without complications
within 4 weeks with only supportive care, but fatigue may
continue for several months longer. Complications that may
occur are generally mild and include hemolytic anemia, mod-
erate thrombocytopenia, aplastic anemia, disseminated intra-
vascular coagulation, thrombotic thrombocytopenic purpura,
or hemolytic uremic syndrome. In rare cases, Guillain-Barr
syndrome or other neurologic complications may occur.77
Hematologic findings resemble those seen in many viral disor-
ders. There may be mild normocytic, normochromic anemia
Figure 28-24 Cytotoxic lymphocyte. Note the granules in the cytoplasm.
and a slight thrombocytopenia. The WBC count is usually
This particular cell is also showing evidence of its motility in the uropod-like
elevated to a range of 11 to 20 109/L with approximately
extension of cytoplasm.
15% of patients having leukocyte counts in excess of 20
109/L.78 The WBC differential shows a relative neutropenia
with a relative and absolute lymphocytosis, possible monocy-
cytotoxic T lymphocytes as well as NK lymphocytes is the pres- tosis, and mild eosinophilia. There is a wide variation in
ence of azure granules (Figure 28-24). lymphocytes, with up to 50% or higher exhibiting reactive
features such as increased cytoplasm, retained basophilia, lob-
ulated or oval nuclei with strands, and clumps of chromatin
INFECTIOUS MONONUCLEOSIS
such as described earlier as effector T cells. The overall lympho-
Although reactive lymphocytes may be seen in other disorders cyte morphology in infectious mononucleosis is reflecting a
such as infections by cytomegalovirus, viral influenza, or hepa- strong T cell immune response to the infection of B cells by
titis A and B, infectious mononucleosis is a more frequent the EBV.
cause. Infectious mononucleosis is associated with the EBV Testing for infectious mononucleosis includes rapid
and has an incidence in the United States of 500 cases per screening tests for the detection of heterophile antibodies,
100,000 annually. It has its highest frequency in young adults, antibodies stimulated by the EBV that cross react with anti-
aged 15 to 24 years.75 The virus is found in body fluids, espe- gens found on sheep and horse red cells. However, not every-
cially saliva, which gives rise to one of its nicknames, the one with infectious mononucleosis will produce heterophile
kissing disease. antibodies, especially children. Definitive testing for EBV
The incubation period of infectious mononucleosis is about infection can be performed for detection of IgM and IgG
3 to 7 weeks. Presenting symptoms include the classic triad of antibodies.77
CHAPTER 28 Nonmalignant Leukocyte Disorders 423

SUMMARY
PHA is a genetic disorder of leukocytes, and the cells can be The lipid storage diseases are a group of disorders each of which
confused with immature neutrophils. An acquired form of hypo- is associated with a specific defect in an enzyme necessary for
segmentation called pseudo-PHA can be seen in some malignant the degradation of lipids. The two lipid storage diseases with
myeloproliferative neoplasms. characteristic macrophage morphology are Gaucher disease and
Hereditary hypersegmentation is rare, but it can be confused with Niemann-Pick disease.
a megaloblastic process. Inherited lymphocyte disorders include DiGeorge syndrome, in
Alder-Reilly anomaly is a manifestation of the mucopolysacchari- which the lack or underdevelopment of the thymus results
dosis characterized by metachromatic granules in leukocytes. in decreased T cells; XLA, in which the lack of a kinase results in
Other inherited leukocyte disorders include CHS, a lethal disorder blocked B-cell development; and two types of SCID. Wiskott-Aldrich
characterized by giant lysosomes in all cells, and MHA, which syndrome is a third inherited disorder affecting both T and B cells.
may or may not have clinical symptoms and is characterized by Reactive changes in granulocytes include a left shift, hyperseg-
leukocyte inclusions similar to Dhle bodies, as well as giant mentation, Dhle bodies, toxic granulation, vacuoles, degranula-
platelets. tion, and cytoplasmic swelling.
CGD of childhood is an inherited disorder of the NADPH oxidase Reactive changes in monocytes include occasional immature
system resulting in neutrophils that are incapable of killing many forms, contorted nuclei, and cytoplasmic evidence of transforma-
microorganisms. tion into macrophages.
LADs are a group of disorders caused by mutations in the genes Reactive changes in lymphocytes include the presence of effector
for adhesive molecules required for cells to migrate from the blood B cells (plasma cells and plasmacytoid lymphocytes), effector T
into the tissues. cells (also known as infectious mononucleosis cells), and occa-
The mucopolysaccharidoses are a group of disorders each of which sional blast forms reflecting blastogenesis.
is associated with a specific defect in an enzyme necessary for
the degradation of GAGs such as heparan sulfate, keratan sulfate, Now that you have completed this chapter, go back and
dermatan sulfate, and chondroitin sulfate. The result is the buildup read again the case studies at the beginning and respond
of partially digested GAGs within macrophages and leukocytes. to the questions presented.

R E V I E W Q UESTIONS
1. Which of the following inherited leukocyte disorders is 4. Which of the following lysosomal storage diseases is char-
caused by a mutation in the lamin B receptor? acterized by macrophages with striated cytoplasm and
a. PHA storage of glucocerebroside?
b. CHS a. Sanfilippo syndrome
c. Alder-Reilly anomaly b. Gaucher disease
d. MHA c. Fabry disease
d. Niemann-Pick disease
2. Which of the following inherited leukocyte disorders is
one of a group of disorders with mutations in nonmuscle 5. The neutrophils in CGD are incapable of producing:
myosin heavy-chain IIA? a. Hydrogen peroxide
a. PHA b. Hypochlorite
b. CHS c. Superoxide
c. Alder-Reilly anomaly d. All of the above
d. MHA
6. Individuals with X-linked SCID have a mutation that
3. Which of the following inherited leukocyte disorders affects their ability to synthesize:
might be seen in Hurler syndrome? a. Deaminase
a. PHA b. Oxidase
b. CHS c. IL-2 receptor
c. Alder-Reilly anomaly d. IL-8 receptor
d. MHA
424 PART V Leukocyte Disorders

7. An absolute lymphocytosis with reactive lymphocytes sug- 9. The expected complete blood count (CBC) results for
gests which of the following conditions? women in active labor would include:
a. DiGeorge syndrome a. High total white blood cell (WBC) count with
b. Bacterial infection increased lymphocytes
c. Parasitic infection b. High total WBC count with a slight shift to the left in
d. Viral infection neutrophils
c. Normal WBC count with increased eosinophils
8. What leukocyte cytoplasmic inclusion is composed of d. Low WBC count with increased monocytes
ribosomal RNA?
a. Primary granules 10. Which of the following is true of the reactive lymphocytes
b. Toxic granules whose cytoplasm has been described as looking like a
c. Dhle bodies flared skirt or fried egg?
d. Howell-Jolly bodies a. They are commonly seen in infectious
mononucleosis.
b. They are effector B cells.
c. They are undergoing blastogenesis.
d. They are characteristic of infectious lymphocytosis.

REFERENCES
1. Zwerger M, Herrmann H, Gaines P, et al: Granulocytic nuclear 14. Cawley JC, Hayhoe FG: The inclusions of the May-Hegglin
differentiation of lamin B receptordeficient mouse EPRO anomaly and Dhle bodies of infection: an ultrastructural
cells. Exp Hematol 36:977-987, 2008. comparison. Br J Haematol 22:491-496, 1972.
2. Hoffmann K, Dreger CK, Olins AL, et al: Mutations in the 15. Stasia MJ, Li XJ: Genetics and immunopathology of chronic
gene encoding the lamin B receptor produce an altered granulomatous disease. Semin Immunopathol 30:209-235,
nuclear morphology in granulocytes (Pelger-Hut anomaly). 2008.
Nat Genet 31:410-414, 2002. 16. Harris ES, McIntyre TM, Prescott SM, et al: The leukocyte
3. Dechat T, Pfleghaar K, Sengupta K, et al: Nuclear lamins: integrins. J Biol Chem 275:23409-23412, 2000.
major factors in the structural organization and function of 17. Bunting M, Harris ES, McIntyre TM, et al: Leukocyte adhesion
the nucleus and chromatin. Genes Dev 22:832-853, 2008. deficiency syndromes: adhesion and tethering defects involv-
4. Hut GJ: ber eine bisher unbekannte familire Anomalie ing 2 integrins and selectin ligands. Curr Opin Hematol 9:30-
der Leukocyten. Klin Wochenschr 2:1264-1266, 1932. 35, 2002.
5. Cohen TV, Kiarmann KD, Sakchaisri K, et al: The lamin B 18. Svensson L, Howarth K, McDowall A, et al: Leukocyte adhe-
receptor under transcriptional control of C/EBP is required sion deficiency-III is caused by mutations in KINDLIN3
for morphological but not functional maturation of neutro- affecting integrin activation. Nat Med 15:306-312, 2009.
phils. Hum Mol Genet 17:2921-2933, 2008. 19. Grimbacher B, Holland SM, Puck JM: Hyper-IgE syndromes.
6. Cunningham JM, Patnaik MM, Hammerschmidt DE, et al: Immunol Rev 203:244-250, 2005.
Historical perspective and clinical implications of the Pelger- 20. Ohga S, Nomura A, Ihara K, et al: Cytokine imbalance in
Hut cell. Am J Hematol 84:116-119, 2009. hyper-IgE syndrome: reduced expression of transforming
7. Aprikyan AA, Liles WC, Park JR, et al: Myelokathexis, a growth factor and interferon genes in circulating activated
congenital disorder of severe neutropenia characterized by T cells. Br J Haematol 121:324-331, 2003.
accelerated apoptosis and defective expression of bcl-x in 21. Miller ME, Oski FA, Harris MB: Lazy leukocyte syndrome.
neutrophil precursors. Blood 95:320-327, 2000. A new disorder of neutrophil function. Lancet 1:665-669,
8. Presentey B: Alder anomaly accompanied by a mutation of 1971.
the myeloperoxidase structural gene. Acta Haematol 75:157- 22. Patrone F, Dallegri F, Rebora A, et al: Lazy leucocyte syn-
159, 1986. drome. Blut 39:265-269, 1979.
9. Kaplan J, DeDomenico I, Ward D: Chdiak-Higashi syn- 23. Marchetti C, Patriarca P, Solero GP, et al: Genetic characteriza-
drome. Curr Opin Hematol 15:22-29, 2008. tion of myeloperoxidase deficiency in Italy. Hum Mutat
10. Tchernev VT, Mansfield TA, Giot L, et al: The Chdiak-Higashi 23:496-505, 2004.
protein interacts with SNARE complex and signal transduc- 24. Cutler CW, Eke P, Arnold RR, et al: Defective neutrophil func-
tion proteins. Mol Med 8:56-64, 2002. tion in an insulin-dependent diabetes mellitus patient. A case
11. Rendu F, Breton-Gorius J, Lebret M, et al: Evidence that report. J Periodontol 62:394-401, 1991.
abnormal platelet functions in human Chdiak-Higashi syn- 25. Osar Z, Samanci T, Demirel GY, et al: Nicotinamide effects
drome are the result of a lack of dense bodies. Am J Pathol oxidative burst activity of neutrophils in patients with poorly
111:307-314, 1983. controlled type 2-diabetes mellitus. Exp Diabesity Res 5:155-
12. Dong F, Li S, Pujol-Moix N, et al: Genotype-phenotype cor- 162, 2004.
relation in MYH9-related thrombocytopenia. Br J Haematol 26. Brck W, Goebel HH, Dienes P: B and T lymphocytes are
130:620-627, 2005. affected in lysosomal disordersan immunoelectron micro-
13. Demeter J, Lelkes G, Nemes L, et al: Familial occurrence scopic study. Neuropathol Appl Neurobiol 17:219-222, 1991.
of the May-Hegglin anomaly: is the accompanying renal 27. Muenzer J: The mucopolysaccharidoses: a heterogeneous
failure part of a new subentity? Ann Hematol 80:368-371, group of disorders with variable pediatric presentations.
2001. J Pediatr 144(5 suppl):S27-S34, 2004.
CHAPTER 28 Nonmalignant Leukocyte Disorders 425
28. Chen M, Wang J: Gaucher disease: review of the literature. barrier permeability induction. Med Hypotheses 70:582-584,
Arch Pathol Lab Med 132:851-853, 2008. 2008.
29. Lim-Melia ER, Kronn DF: Current enzyme replacement 51. Anzai M, Maezawai R, Ohara T, et al: Systemic lupus erythe-
therapy for the treatment of lysosomal storage diseases. matosus associated with facial edema, overproduction of
Pediatr Ann 38:448-455, 2009. interleukin-5, and eosinophilia. J Clin Rheumatol 14:361-362,
30. Lieberman RL, Daquino JA, Ringe D, et al: Effects of pH and 2008.
iminosugar pharmacological chaperones on lysosomal glyco- 52. Gleich GF, Leiferman KM: The hypereosinophilic syndromes:
sidase structure and stability. Biochemistry 48:4816-4827, current concepts and treatments. Br J Haematol 145:271-285,
2009. 2009.
31. Sharma P, Khurana N, Singh T: Pseudo-Gaucher cells in Hb 53. Freeman GL: Syndromes associated with eosinopenia. Allergy
E disease and thalassemia intermedia. Hematology 12:457- 53:331-333, 1998.
459, 2007. 54. Grattan CE, Dawn G, Gibbs S, et al: Blood basophil numbers
32. Lee RE, Ellis LD: The storage cells of chronic myelogenous in chronic ordinary urticaria and healthy controls: diurnal
leukemia. Lab Invest 24:261-264, 1971. variation, influence of loratadine and prednisolone and
33. Carrington PA, Stevens RF, Lendon M: Pseudo-Gaucher cells. relationship to disease activity. Clin Exp Allergy 33:337-341,
J Clin Pathol 45:360, 1992. 2003.
34. Kolodny EH: Niemann-Pick disease. Curr Opin Hematol 7:48- 55. Dameshek W: The appearance of histiocytes in the peripheral
52, 2000. blood. Arch Intern Med 47:968-985, 1931.
35. Schuchman EH: The pathogenesis and treatment of acid 56. Maldonado JE, Hanlon DG: Monocytosis: a current appraisal.
sphingomyelinase-deficient Niemann-Pick disease. J Inherit Mayo Clinic Proc 40:248-259, 1965.
Metab Dis 30:654-663, 2007. 57. Chakraborty A, Blum RA, Cutler DL, et al: Pharmacoimmu-
36. Carstea ED, Morris JA, Coleman KG, et al: Niemann-Pick C1 nodynamic interactions of interleukin-10 and prednisone in
disease gene: homology to mediators of cholesterol homeo- healthy volunteers. Clin Pharmacol Ther 65:304-318, 1999.
stasis. Science 277:228-231, 1997. 58. Nockher WA, Wiemer J, Scherberich JE: Haemodialysis
37. Naureckiene S, Sleat DE, Lackland H, et al: Identification of monocytopenia: differential sequestration kinetics of CD14+
HE1 as the second gene of Niemann-Pick C disease. Science CD16+ and CD14++ blood monocyte subsets. Clin Exp
290:2298-2301, 2000. Immunol 123:49-55, 2001.
38. Lloyd-Evans E, Morgan AJ, He X, et al: Niemann-Pick 59. Savard M, Gosselin J: Epstein-Barr virus immunosuppression
disease type C1 is a sphingosine storage disease that causes of innate immunity mediated by phagocytes. Virus Res
deregulation of lysosomal calcium. Nat Med 14:1247-1255, 119:134-145, 2006.
2008. 60. Ponder E, Ponder RV: The cytology of the polymorphonu-
39. McLean-Tooke A, Spickett GP, Gennery AR: Immunodefi- clear leukocyte in toxic conditions. J Lab Clin Med 28:316-321,
ciency and autoimmunity in 22q11.2 deletion syndrome. 1942.
Scand J Immunol 66:1-7, 2007. 61. Prokocimer M, Potasman I: The added value of peripheral
40. Markert ML, Devlin BH, Alexieff MJ, et al: Review of 54 blood cell morphology in the diagnosis and management of
patients with complete DiGeorge anomaly enrolled in proto- infectious diseasespart 1: basic concepts. Postgrad Med J
cols for thymus transplantation: outcome of 44 consecutive 84:579-585, 2008.
transplants. Blood 109:4539-4547, 2007. 62. Abernathy MR: Dhle bodies associated with uncomplicated
41. Lindvall JM, Blomberg KE, Vliaho J, et al: Brutons tyrosine pregnancy. Blood 27:380-385, 1966.
kinase: cell biology, sequence conservation, mutation spec- 63. Schofield KP, Stone PC, Beddall AC, et al: Quantitative cyto-
trum, siRNA modifications, and expression profiling. Immunol chemistry of the toxic granulation blood neutrophil. Br J
Rev 203:200-215, 2005. Haematol 53:15-22, 1983.
42. Minegishi Y, Rohrer J, Conley ME: recent progress in 64. Kabutomori O, Iwatani Y, Kanakura Y: Toxic granulation neu-
the diagnosis and treatment of patients with defects in trophils and C-reactive protein. Arch Intern Med 160:3326-
early B-cell development. Curr Opin Pediatr 11:528-532, 3327, 2000.
1999. 65. Al-Gwaiz LA, Babay HH: The diagnostic value of absolute
43. Hershfield MS: New insights into adenosine-receptor neutrophil count, band count and morphologic changes in
mediated immunosuppression and the role of adenosine in neutrophils in predicting bacterial infections. Med Princ Pract
causing the immunodeficiency associated with adenosine 16:344-347, 2007.
deaminase deficiency. Eur J Immunol 35:25-30, 2005. 66. Kabutomori O, Kanakura Y, Watani YI: Induction of toxic
44. Uribe L, Weinberg KI: X-linked SCID and other defects of granulation in neutrophils by granulocyte colony-stimulating
cytokine pathways. Semin Hematol 35:299-309, 1998. factor. Eur J Haematol 69:187-188, 2002.
45. Notarangelo LD, Miao CH, Ochs HD: Wiskott-Aldrich syn- 67. von Gunten A, Simon HU: Autophagic-like cell death in neu-
drome. Curr Opin Hematol 15:30-36, 2008. trophils induced by autoantibodies. Autophagy 3:67-68, 2007.
46. Welte K, Zeidler C: Sever congenital neutropenia. Hematol 68. Davidson RJ, McPhie JL: Cytoplasmic vacuolation of periph-
Oncol Clin North Am 23:307-320, 2009. eral blood cells in acute alcoholism. J Clin Pathol 33:1193-
47. Skokowa J, Fobiwe JP, Dan L, et al: Neutrophil elastase is 1196, 1980.
severely down-regulated in severe congenital neutropenia 69. Holley TR, Van Epps DE, Harvey RL, et al: Effect of high doses
independent of ELA2 or HAX-1 mutations but dependent on of radiation on human neutrophil chemotaxis, phagocytosis
LEF-1. Blood 114:3044-3051, 2009. and morphology. Am J Pathol 75:61-72, 1974.
48. Filley WV, Holley KE, Kephart GM, et al: Identification by 70. Centers for Disease Control and Prevention, Tickborne
immunofluorescence of eosinophil granule major basic Rickettsial Diseases Working Group: Diagnosis and manage-
protein in lung tissues of patients with bronchial asthma. ment of tickborne rickettsial diseases: Rocky Mountain
Lancet 2:11-16, 1982. spotted fever, ehrlichioses, and anaplasmosisUnited States.
49. Simon D, Braathen LR, Simon HU: Eosinophils and atopic MMWR Recomm Rep 55(RR-4):1-27, 2006.
dermatitis. Allergy 59:561-570, 2004. 71. Centers for Disease Control and Prevention: Ehrlichiosis.
50. Mingomataj EC: Eosinophil-induced prognosis improvement Available at: http://www.cdc.gov/ticks/diseases/ehrlichiosis.
of solid tumors could be enabled by their vesicle-mediated Accessed June 20, 2010.
426 PART V Leukocyte Disorders

72. Centers for Disease Control and Prevention: Anaplasmosis. 76. Hurt C, Tammaro D: Diagnostic evaluation of mononucleosis-
Available at: http://www.cdc.gov/ticks/diseases/anaplasmosis/ like illnesses. Am J Med 120:911.e1-911.e8, 2007.
index.html. Accessed June 20, 2010. 77. Luzuriaga K, Sullivan JL: Infectious mononucleosis. N Engl J
73. Paddock CD, Childs JE: Ehrlichia chaffeensis: a prototypical Med 362:1993-2000, 2010.
emerging pathogen. Clin Microbiol Rev 16:37-64, 2003. 78. Gross TG: Infectious mononucleosis and other Epstein-Barr
74. Gallen JI: Neutrophil specific granules, a fuse that ignites the virus-related disorders. In Greer JP, Foerster J, Rodgers GM,
inflammatory response. Clin Res 32:320-328, 1984. et al, editors: Wintrobes clinical hematology, ed 12, Philadel-
75. Crawford DH, Macsween KF, Higgins CD, et al: A cohort phia, 2009, Lippincott Williams & Wilkins.
study among university students: identification of risk factors
for Epstein-Barr virus seroconversion and infectious mono-
nucleosis. Clin Infec Dis 43:276-282, 2006.
Introduction to
Leukocyte Neoplasms
29
Peter D. Emanuel

OUTLINE OBJECTIVES
General Characteristics of After completion of this chapter, the reader will be able to:
Leukocyte Neoplasms
Etiology of Leukocyte 1. Name two viruses that play a pathogenic role in 7. Explain why localized treatments are rarely used for
Neoplasms lymphoid malignancies. leukocyte neoplasms.
Classification Schemes for 2. Define oncogene and tumor suppressor gene and 8. Describe examples of supportive care therapies that
Leukocyte Neoplasms explain the difference between them, including the have been developed from molecular biology tech-
Chromosomal mechanisms by which each produces cancer. niques, including their functions and purposes.
Abnormalities in 3. Explain why oncogenes are described as acting in a 9. Name two examples of targeted therapeutics for
Hematologic Neoplasms dominant fashion. chronic myelogenous leukemia and explain their
Oncogenes 4. Give two examples of oncogenes and two examples methods of action.
Tumor Suppressor Genes of tumor suppressor genes. 10. Describe the differences between small molecule
Other Genetic Changes
5. Describe how genetic abnormalities can affect inhibitors and monoclonal antibodies.
Molecular Pathways
molecular pathways within cells. 11. Name three sources of hematopoietic stem cells.
Perturbed by Cellular
Transformation 6. Identify the two basic subgroups of chemotherapeu- 12. Compare and contrast allogeneic and autologous
Therapy for Leukocyte tic agents. bone marrow transplants.
Neoplasms
Chemotherapy
Radiation Therapy
Supportive Therapy
Targeted Therapy
Stem Cell Transplantation

GENERAL CHARACTERISTICS OF ETIOLOGY OF LEUKOCYTE NEOPLASMS


LEUKOCYTE NEOPLASMS
For most leukocyte neoplasms, causes directly related to the
Most malignancies of the hematopoietic system are acquired development of the malignancy are unknown. There are,
genetic diseases, which means that most patients are not born however, a few exceptions. Environmental toxins can induce
with the illness but acquire it sometime later. Most leukocyte genetic changes, as discussed later in this chapter, leading to a
malignancies are not localized but rather are systemic at the malignancy phenotype. Environmental exposures known to
initiation of the malignant process. The bone marrow, in lead to hematopoietic malignancies include radiation expo-
which the leukemias arise, and the lymphatic system, in which sure, as experienced by survivors of atomic explosions, and
most lymphomas originate, access passages throughout the exposure to organic solvents, such as benzene. There are two
body. A single leukemia cell arising in the marrow can pass types of lymphoid malignancies in which viruses may play a
into the bloodstream and travel to any and all locations of the pathogenetic role. The Epstein-Barr virus has been implicated
body. With rare exceptions, most treatments for leukocyte neo- as an etiologic factor in the development of Burkitt non-
plasms given with curative intent are not localized, such as Hodgkin lymphoma. Similarly, human T-cell lymphotropic
radiation or surgery, but must by nature be systemic-type virus type 1 (HTLV-1) is the likely cause of adult T-cell leukemia/
treatments. lymphoma. As discussed further in this chapter, there are some

427
428 PART V Leukocyte Disorders

known familial cancer predisposition syndromes. In addition, associated with consistently recurring chromosomal transloca-
as more cancer survivors live longer, it is clear that some alkyl- tions. This 2001 WHO classification of hematologic malignan-
ating agents and other forms of chemotherapy used to treat cies has undergone a recent revision.2
various forms of cancer can induce deoxyribonucleic acid
(DNA) damage in hematopoietic cells, leading to hematologic
CHROMOSOMAL ABNORMALITIES IN
malignancies.
HEMATOLOGIC NEOPLASMS
In part because of the ease of access to bone marrow compared
CLASSIFICATION SCHEMES FOR
with other organs of the body, researchers have learned a great
LEUKOCYTE NEOPLASMS
deal about the biology of cancer by studying hematopoietic
The French-American-British (FAB) classification of the acute neoplasms. The study of chromosomal translocations in hema-
leukemias was devised in the 1970s and 1980s. The FAB tologic malignancies has taught how a single mutation, or
schemas were based largely on morphologic characteristics and series of mutations in stepwise fashion, can lead to malignant
relied heavily on examination of routine histologic stain prepa- transformation by disrupting the molecular machinery of the
rations to distinguish lymphoid neoplasms from myeloid cell. The first two genetic lesions found in any kind of human
neoplasms (Figure 29-1). Although these types of diagnostic cancer were identified as chromosomal translocations occur-
criteria have not been abandoned, pathologists are now moving ring in leukocyte malignancies. These were the t(9;22) trans
toward more precise classification of many of the leukocyte location in chronic myelogenous leukemia (CML)3 and the
neoplasms based on recurring chromosomal and genetic t(8;14) translocation in Burkitt lymphoma.4
lesions found in many patients. These lesions are related to
disruptions of oncogenes, tumor suppressor genes, and other
ONCOGENES
regulatory elements that control proliferation, maturation,
apoptosis, and other vital cell functions. In 2001 the World Oncogenes originally were identified as genes that carried
Health Organization (WHO) published new classification rapidly transforming retroviruses derived from normal cellular
schemes for nearly all of the tumors of hematopoietic and homologues, proto-oncogenes. The oncogene definition has
lymphoid tissues.1 In some cases WHO melded the older mor- evolved to specify genes that cause dominant-acting cancer
phologic schemes with the newer schemes. For instance, in the mutations, regardless of whether they are derived from a retro
WHO classification scheme for acute myeloid leukemias virus. The typical proto-oncogene codes for a protein involved
(AMLs) there are some remnants of the old FAB classification, in normal cell cycle regulation. Regulation proteins provide the
but new classifications were introduced for leukemias signal transduction that carries messages about cell division

A B

Lymphoblasts: Myeloblasts:

2-3 times the diameter of lymphocytes 3-5 times the diameter of lymphocytes
Scant blue cytoplasm Moderate gray cytoplasm
Uniform coarse chromatin Uniform fine chromatin
Inconspicuous nucleoli 2 or more prominent nucleoli
Possible Aer rods
Figure 29-1 A, Lymphoblasts (bone marrow, Wright stain, 500). Cells have a diameter two to three times the normal lymphocyte diameter, scant blue
cytoplasm, coarse chromatin, deeper staining than myeloblasts, and inconspicuous nucleoli. B, Myeloblasts (bone marrow, Wright stain, 500). Cells have a
diameter three to five times the lymphocyte diameter, moderate gray cytoplasm, uniform fine chromatin, two or more prominent nucleoli, and possibly Aer
rods.
CHAPTER 29 Introduction to Leukocyte Neoplasms 429

and maturation (differentiation) from outside the cell to the formation of oncogenes or loss of tumor suppressor genes has
nucleus. Mutations of these genes may form oncogenes that on the proliferation and differentiation of hematologic tissues.
disrupt normal cell cycle processes. Specialists recognize several mechanisms, including blocked dif-
Most chromosomal translocations in leukemias involve ferentiation, transcriptional repression, disruptions of cell signaling,
oncogenes. The dominant transforming oncogene is able to progression, and apoptosis. The t(15;17) translocation found in
alter the gene product and transform the cell into a malignant acute promyelocytic leukemia (APL), which fuses the PML gene
phenotype, even in the presence of a residual normal allele. to the RARA (retinoic acid receptor alpha) gene, clearly results
Even the afore-mentioned two examples, CML and Burkitt lym- in a state of arrested differentiation, because RARA-induced dif-
phoma, t(9;22) and t(8;14), involve oncogenes that are acti- ferentiation is inhibited. Treating patients with APL with phar-
vated when brought into proximity with their new partners on macologically high doses of all-trans retinoic acid can overcome
fusion genes. In the case of CML, the ABL proto-oncogene on this block and permit APL cells to differentiate into normal
chromosome 9 is activated when fused to the BCR component neutrophils. In so doing, the APL cells lose their leukemic
of chromosome 22. In the case of Burkitt lymphoma, the MYC potential.
proto-oncogene on chromosome 8 is fused with the immuno- Other chromosomal abnormalities involve transcriptional
globulin heavy chain locus on chromosome 14. Although repression of DNA and condensation abnormalities of chroma-
oncogenic transformation was first identified by karyotypic tin, such as those involved in the core-binding factor leukemic
analyses, molecular biologic techniques have evolved rapidly subtypes of AML. A similar example on the lymphoid side of
over the last three decades, so that more genetic translocations hematopoietic neoplasms are the chromosomal translocations
are being identified that create novel fusion genes invisible at involving the BCL6 gene. Normal BCL6 encodes for a transcrip-
the cytogenetic level. tional repressor responsible for recruiting the histone deacetylase
complex, which regulates germinal center formation in lymph
nodes. The mutation of BCL6 leads to overexpression of this
TUMOR SUPPRESSOR GENES
normal protein so that DNA is excessively repressed, which in
Tumor suppressor genes are so named because they code for this case prevents lymphocytes from progressing beyond the
proteins that resist malignant transformation. These genes do germinal center stage of development.
not act in a dominant fashion as in the case of oncogenes; Since the initial identification of the BCR/ABL fusion gene
rather, cells are transformed into a malignant phenotype only in CML, there are now many other examples of genetic abnor-
after both alleles of these genes have been lost or otherwise malities in myeloid malignancies in which the abnormality
inactivated, the so-called two-hit mechanism proposed by leads to disruption of cell signaling, often by way of activation
Knudson.5 Although tumor suppressor genes are harder to of kinase cascades. FLT3 codes for a tyrosine kinase receptor
isolate and identify, numerous such genes have now been iden- preferentially expressed on hematopoietic stem cells that medi-
tified, and many have been found to be associated with auto- ates proliferation and differentiation. A unique mutation
somal dominant familial cancer predisposition syndromes. resulting in an internal tandem duplication leads to constitu-
Some well-known examples include the RB1 tumor suppressor tive activation of this pathway (i.e., always turned on) in
gene involved in familial retinoblastoma, the TP53 gene in many forms of AML and other hematopoietic malignancies.
Li-Fraumeni syndrome, the WT1 gene in Wilms tumor, and the Other examples of gene mutations that alter kinase cascades
NF1 gene in familial neurofibromatosis type 1. Perhaps more in myeloid or lymphoid cells are c-KIT, NOTCH, JAK2,
importantly, many of these tumor suppressor genes and their and RAS.
protein products are altered in many sporadic cancers, includ- Many cyclin-dependent kinases are altered in lymphoid malig-
ing hematologic cancers. nancies. The cyclin-dependent kinases tightly regulate cell cycle
progression through the synthesis, proteolysis, and phosphory-
lation of cyclins.
OTHER GENETIC CHANGES
Finally, another important molecular signaling pathway in
Beyond oncogenes and tumor suppressor genes, researchers cells involves programmed cell death, or apoptosis. This vital
have found genes that are involved in DNA repair mechanisms. process allows organisms to eliminate redundant, damaged,
Mutations in these genes do not lead to cellular transformation aged, or infected cells. In the hematopoietic environment,
by altering signaling pathways, but rather lead to genetic insta- apoptosis is essential to contain and control the massive expan-
bility and increased mutation rates. Examples of such genes are sion that the hematopoietic system is capable of generating at
the DNA mismatch repair genes MLH1 and MSH2. Another times of stress, infection, or hemorrhage. Caspases are a family
example is the Fanconi anemia gene, FA, which is important of proteases that participate in the apoptotic cascade triggered
for maintaining genomic stability in hematopoietic tissues. in response to proapoptotic signals. The culmination of this
apoptotic cascade is cellular disassembly. The BCL family con-
tains many genes, some of which are proapoptotic and some
MOLECULAR PATHWAYS PERTURBED BY
of which are antiapoptotic. Many of the various forms of non-
CELLULAR TRANSFORMATION
Hodgkin lymphoma seem to involve disruptions of BCL2,
Regardless of the type of chromosomal or genetic abnormality, BCL6, BCL10, or other members of the caspase and BCL family
clinicians understand the molecular consequences that the of genes comprising the apoptotic cascade.
430 PART V Leukocyte Disorders

The list of chromosomal and molecular aberrations known increase in killing of cells with a further increase in dosage.
to occur in the various leukocyte neoplasms continues to grow Examples are L-asparagine amidohydrolase (G1 phase), antime-
on an almost daily basis. Indexing this list is far beyond the tabolites such as methotrexate (S phase), and vinca alkaloids
scope of this chapter, but some condensed lists are provided (M phase).
in Chapter 31. More complete indices can be found in other Chemotherapeutic agents affect both neoplastic and normal
publications such as the WHO reclassification scheme,1 its cells. The effect is most pronounced on rapidly dividing cells,
2008 revision, or other hematology textbooks.6-10 such as those of the mucosa of the gastrointestinal tract and
the bone marrow. This limits the dosage and usually deter-
mines the maximum tolerated dose for a patient. Chemother-
THERAPY FOR LEUKOCYTE NEOPLASMS
apy agents are categorized in Table 29-1.
The various forms of therapy available today for leukocyte
neoplasms can be roughly divided into the following catego- Alkylating Agents
ries: chemotherapy, radiation therapy, supportive therapy, targeted The mechanism of action of alkylating agents is to ionize
therapy, and stem cell transplantation. In contrast to many solid within cells, forming highly reactive free radicals that damage
tumors, numerous hematologic malignancies now have cure DNA. These agents act on cells in any phase of the cell cycle.
rates that are substantially higher than they were two or three They include nitrogen mustard, cyclophosphamide, chloram-
decades ago. Many new and exciting therapies that are less toxic bucil, busulfan, and melphalan.
are now under development or are already employed in patient
settings. These therapies are bringing more optimism to the Plant Alkaloids
care of patients with leukocyte neoplasms than ever before. Plant alkaloids affect microtubules and interrupt the process of
Selection of the best therapy must start, however, with an mitotic spindle formation during the metaphase stage of
accurate diagnosis. Even the most effective therapies do not mitosis. These agents include vincristine and vinblastine.
work if they are applied in the wrong circumstances.
Curative treatment strategies are a realistic goal for patients Antitumor Antibiotics
with Hodgkin lymphoma, CML, hairy cell leukemia, and some Compounds derived from living microorganisms are termed
forms of non-Hodgkin lymphoma, and for children with acute antibiotics, and some have antitumor effects. Antibiotics inhibit
lymphoblastic leukemia. Cure may be attainable in other patients the synthesis of ribonucleic acid (RNA) or DNA and interfere
with acute lymphoblastic or myeloid leukemia, and long-term with the G2 phase of the cell cycle. Commonly used tumor
remissions may be achievable in adults with multiple myeloma. antibiotics include daunorubicin and doxorubicin.
For patients with other leukocyte neoplasms such as mantle
cell lymphoma, chronic lymphocytic leukemia, or a therapy- Antimetabolites
related leukemia, cure remains elusive, and therapy must be Antimetabolites interfere with the normal functions of various
directed more toward attaining remissions or providing sup- essential metabolites. Examples are methotrexate, folate anta
portive care. gonists, and the purine analogues such as 6-mercaptopurine
and 6-thioguanine, which most often affect cells in S phase.
Chemotherapy
Chemotherapy is oral or parenteral cancer treatment with com- Glucocorticoids
pounds that possess antitumor properties. The methods of The synthetic or natural steroids include compounds such as
action of the chemotherapy drugs vary considerably. Chemo- hydrocortisone, prednisone, dexamethasone, and predniso-
therapy agents can be classified in two ways: by their effects on lone. Steroids have a lympholytic effect and affect nonpro
the cell cycle and by their biochemical mechanism of action. liferating cells and those in cycle. Protein synthesis and mitosis
Some chemotherapy drugs can affect cells only in specific also may be inhibited.
phases of the cell cycle (phase specific), whereas other drugs
act without regard to the cell cycle (phase nonspecific) and Radiation Therapy
affect cells during any phase of the cell cycle. Agents in this The effectiveness of x-rays against Hodgkin and non-Hodgkin
latter category usually have a linear dose-response curve (i.e., lymphomas was described shortly after radiations discovery.
the higher the dose, the more cells are killed). There are two Radiation kills cells by producing unstable ions that damage
subgroups: DNA and may cause instant or delayed death of the cell. The
1. Cycle-specific agents, which kill cells that are moving toxic effects of radiotherapy can occur during therapy or much
through the cell cycle, regardless of whether the cells are in later. Complications can be reduced through the use of com-
G1, G2, S, or M phase (e.g., alkylating agents, cisplatin). bined anterior and posterior treatment ports and the applica-
2. Cycle-nonspecific agents, which kill nondividing cells or tion of maximal shielding techniques to prevent damage to
cells in the resting state (e.g., steroids and antitumor normal tissues. The hematopoietic system, the gastrointestinal
antibiotics). tract, and the skin are most often affected during radiotherapy.
Phase-specific agents are effective only if present during The toxic effects are usually reversible when radiation is
a certain phase in the cell cycle (see Chapters 31 and 32). stopped. The epithelium of the entire gastrointestinal tract is a
Within a certain dosage range, agents in this category show no rapidly dividing cellular system that is very sensitive to
CHAPTER 29 Introduction to Leukocyte Neoplasms 431

TABLE 29-1 Chemotherapy Agents


Agent Other Names Uses Toxic Effects
Alkylating Agents
Busulfan Myleran CML, pretransplantation Myelosuppression, infertility
Cyclophosphamide Cytoxan, Neosar Lymphoma, MM, ALL, pretransplantation Marrow suppression, N&V, cystitis
Nitrogen mustard Mechlorethamine Hodgkin lymphoma, NHL Myelosuppression, N&V, infertility
Chlorambucil Leukeran CLL, Waldenstrm macroglobulinemia, Myelosuppression, hair loss
NHL, Hodgkin disease
Melphalan Alkeran MM Myelosuppression
Carmustine BCNU Hodgkin disease, NHL, MM Myelosuppression
Dacarbazine DTIC Hodgkin disease Myelosuppression, N&V

Plant Alkaloids
Vincristine Oncovin ALL, NHL, Hodgkin disease, CLL, MM Neurotoxicity, hair loss
Vinblastine Velban ALL, NHL, Hodgkin disease, CLL, MM Myelosuppression
Etoposide VP-16 NHL, pretransplantation Myelosuppression, hair loss

Antitumor Antibiotics
Daunorubicin Daunomycin AML, ALL Myelosuppression, cardiotoxicity, N&V, hair loss
Doxorubicin Adriamycin ALL, AML, Hodgkin disease, NHL, CLL, MM Myelosuppression, cardiotoxicity, N&V, hair loss
Bleomycin Bleo Hodgkin lymphoma, NHL Lung toxicity, gastrointestinal toxicity
Idarubicin Idamycin AML Myelosuppression

Antimetabolites
Methotrexate Amethopterin, MTX ALL, NHL Myelosuppression, gastrointestinal toxicity
Ara-C Cytosine arabinoside, AML, NHL, Hodgkin disease Myelosuppression, gastrointestinal toxicity,
cytarabine hair loss
Mercaptopurine 6-MP ALL, CML Hepatotoxicity, myelosuppression
Thioguanine 6-TG AML Myelosuppression
Pentostatin 2-Deoxycoformycin Hairy cell leukemia, CLL, lymphomas Neurotoxicity, myelosuppression
Fludarabine CLL, lymphomas, Waldenstrm Neurotoxicity, myelosuppression
macroglobulinemia
2-CDA CDA, 2-chlorodeoxyadenosine, Hairy cell leukemia, CLL, lymphomas, AML, Neurotoxicity, myelosuppression
2-cladribine Waldenstrm macroglobulinemia

Glucocorticoids
Prednisone ALL, CLL Fluid retention, muscle weakness
Methylprednisolone Hodgkin disease, NHL, AMM Fluid retention, muscle weakness
Hydrocortisone Hodgkin disease, NHL, AMM Fluid retention, muscle weakness
Dexamethasone Decadron MM Fluid retention, muscle weakness

Others
Hydroxyurea Hydrea CML, PV, AMM Leukopenia, N&V
Asparaginase L-Asparagine amidohydrolase ALL, refractory NHL Nephrotoxicity, N&V
Cisplatin Cis-platinum Solid tumors, Hodgkin disease, NHL Nephrotoxicity, ototoxicity
Procarbazine Hodgkin disease, NHL Myelosuppression

ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; AMM, agnogenic myeloid metaplasia; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia;
MM, multiple myeloma; NHL, non-Hodgkin lymphoma; N&V, nausea and vomiting; PV, polycythemia vera.

radiation. There may be drying up of saliva and loss of taste. can cause marrow suppression, sometimes lowering blood
If the stomach is irradiated, anorexia, nausea, and vomiting counts to the life-threatening range.
may occur. Intestinal irradiation may result in malabsorption
and diarrhea. Irradiated skin becomes erythematous and Supportive Therapy
tender. Permanent loss of body hair and hyperpigmentation Numerous substances that are naturally produced in the
also may occur in irradiated areas. Spinal and pelvic irradiation human body have now been created in the laboratory using
432 PART V Leukocyte Disorders

cloning techniques. Several are commercially produced and antibodies for targeted therapeutic strategies. Rituximab specifi-
have been cleared by the Food and Drug Administration cally targets the CD20 antigen present on many non-Hodgkin
(FDA) for general use in the supportive care of cancer patients, lymphoma cells. When the antibody binds the lymphoma cell,
particularly patients with hematologic malignancies. Colony- complement is activated, and the cell lyses. Other possible
stimulating factors (CSFs), a class of cytokines, normally act scenarios leading to cell death with the use of monoclonal
in the bone marrow microenvironment to stimulate blood antibodies include antibody-mediated cellular cytotoxicity and
cell formation. Erythropoietin is the main stimulatory CSF stimulation of apoptosis.10 In contrast to small-molecule inhib-
responsible for red blood cell formation. Normal erythropoi- itors, monoclonal antibodies generally must be delivered intra-
etin and a long-acting formulation of this molecule are avail- venously or subcutaneously. Monoclonal antibodies such as
able to aid in the care of cancer patients with anemia induced rituximab have evolved from the original monoclonal antibod-
by chemotherapy. Similarly, granulocyte colony-stimulating ies that were derived from mice. Rituximab represents a human-
factor (G-CSF) and granulocyte-macrophage colony-stimulating ized form of an antibody designed or modified to be more
factor (GM-CSF) are used to expand rapidly the number of human so that the patients immune system will not raise an
mature neutrophils capable of fighting infection within the antibody against the monoclonal antibody. Additional modi-
body. Recombinant forms of G-CSF and GM-CSF and a long- fications are now being developed, such as radioactively labeled
acting form of G-CSF are now FDA cleared and are commonly monoclonal antibodies that can carry a killing dose of radio-
employed to support cancer patients with chemotherapy- activity directly to the mutated cell.
induced neutropenia. CSFs not only have enabled physicians
to improve patients quality of life, but also have allowed more Stem Cell Transplantation
efficient and effective delivery of chemotherapy regimens by As more has been learned about hematopoietic stem cells, the
preventing delays of or dosage reductions in chemotherapy therapeutic method of bone marrow transplantation has evolved
courses owing to low blood counts. On the other hand, use of to be more aptly termed stem cell transplantation, because a
erythropoietin support has recently become restricted because variety of different sources in addition to bone marrow can be
of reports of possible higher tumor relapse rates and shortened used to obtain hematopoietic stem cells. Along with bone
survival. marrow, peripheral blood and umbilical cord blood are rich
sources. Regardless of the source, hematopoietic stem cells are
Targeted Therapy considered adult stem cells, even when they come from umbili-
As more has been learned about the specific genetic lesions that cal cord blood, as opposed to embryonic stem cells, which are
cause cancers, researchers have worked to develop targeted the subject of considerable ethical debate. Stem cell transplan-
therapies that act specifically on the tumor cell and leave tation still remains an expensive and rigorous treatment
normal cells untouched.11 As a result of these advances in alternative.
translational research, cancer therapy is realizing the dream of When the decision to transplant has been made and a
targeted therapeutics and is moving away from nonspecific donor has been found, an extensive hospital stay is usually
therapies such as chemotherapy and radiation. The Philadel- required. The pretransplantation conditioning regimen uses
phia chromosomal abnormality, t(9;22) associated with CML high-dose therapy to kill the patients cancer cells and bone
was first reported in 1960. The Philadelphia chromosome marrow cells. This regimen reduces the bodys immunity to
results from the juxtaposition of two genes, BCR and ABLl, and dangerously low levels and necessitates special protective isola-
the resulting fusion protein has elevated tyrosine kinase activ- tion. Granulocyte counts approaching 0 are commonly seen
ity. It took until almost the year 2000 to develop an agent that immediately before and after transplantation. After the infu-
specifically targets the product of the Philadelphia chromo- sion of donor stem cells, the recipient remains in a severely
some. Imatinib mesylate (Gleevec) is a tyrosine kinase inhibitor immunosuppressed condition for 2 weeks or longer. Strict iso-
that inhibits the kinase activity of all proteins that contain ABL. lation at this point is crucial. Prophylactic antibiotics and intra-
Imatinib is not specific solely for the BCR/ABLl tyrosine kinase; venous nutrition are also essential to keep the patient alive
it also inhibits the tyrosine kinase activity of the c-KIT and the until the marrow engrafts. The return of granulocytes, reticulo-
platelet-derived growth factor receptor. It is through its activity cytes, and platelets to normal levels is monitored closely in the
on these other receptors that imatinib has found usefulness peripheral blood. Hematology laboratory evaluation and man-
against other tumors as a targeted therapeutic. In CML, agement of red blood cell and platelet transfusions are crucial
however, imatinib has quickly become the standard treatment components of stem cell transplantation. After the patients
for patients newly diagnosed with the disease who are not release from the hospital, the blood counts, along with bone
candidates for immediate transplant (see Chapter 34). Resis- marrow aspirate and core biopsy specimens, continue to be
tance to imatinib has developed in some CML cells, and monitored to measure the progress of engraftment of the
second-generation, more powerful kinase inhibitors, such as donor stem cells.
dasatinib and nilotinib, have now been cleared by the FDA. Stem cell transplants for treatment of malignant disease
Small-molecule inhibitors such as imatinib are typically avail- have come from donors of three general types: (1) an identical
able in oral form and have relatively few side effects. twin donor (syngeneic transplant), (2) a donor genetically
Rather than utilizing small-molecule inhibitors, treatment different from the recipient (allogeneic transplant), or (3)
of lymphoid neoplasms has relied more on monoclonal the patients own marrow or peripheral blood stem cells
CHAPTER 29 Introduction to Leukocyte Neoplasms 433

ALLOGENEIC AUTOLOGOUS

Donor (related Recipient


or unrelated) Treatment (patient)

1 Peripheral blood 1 Peripheral blood


stem cells stem cells collected
collected or bone or bone marrow
marrow harvested harvested

Blood/marrow Blood/marrow
2 processing; 2 processing and
may include storage; may
T-cell depletion include purging

Blood/
3 4 marrow 3 4 Blood/marrow
infusion infusion

Conditioning: Conditioning:
High-dose High-dose
cyclophosphamide cyclophosphamide
Total body irradiation Total body irradiation
Recipient Recipient Recipient Recipient
(patient) (patient)
Figure 29-2 Peripheral blood stem cell/bone marrow transplantation protocols.

(autologous transplant) (Figure 29-2). Syngeneic transplants disease. Skin lesions, joint contractures, chronic hepatitis, mal-
are most desirable because of the perfect match of cells. For absorption, and chronic obstructive pulmonary disease are
obvious reasons, however, they are rare, and they are not dis- frequent features of the chronic GVHD syndrome. Clinically
cussed further in this chapter. significant GVHD is associated with a fatality risk 25 times
higher than that in patients without GVHD.
Allogeneic Transplantation T-cell depletion of donor bone marrow is the most effective
Most stem cell donors are genetically different from the recipi- means of preventing acute and chronic GVHD, but this benefit
ent. The intent is to match as many of the human leukocyte has been offset by the substantial increase in the risk of leuke-
antigens (HLAs) as possible. Within any given family, there can mic relapse and infections. There is considerable clinical evi-
be only four HLA haplotypes (two from the mother and two dence that allogeneic grafts lower the risk of leukemic relapse.
from the father), and there is one chance in four that a sibling This antileukemia effect is most pronounced in the presence of
of the patient will be HLA identical. In addition to HLA- chronic GVHD.
identical grafts, grafts from HLA-mismatched donors within
families have been used. Autologous Transplantation
A major complication of an allogeneic marrow graft is the In autologous stem cell transplantation, cells are harvested
immunologic reaction of donor T cells against the tissues of from the patient and, after conditioning, transplanted back.
the recipient, which results in graft-versus-host disease (GVHD). Stem cells harvested during remission, presumably contami-
Two forms of GVHD are recognized: acute and chronic. Acute nated with malignant cells, are purged in vitro through the use
GVHD develops in the immediate posttransplantation period of antileukemic monoclonal antibodies or cytotoxic drugs.
or shortly thereafter. It is characterized by a skin rash, liver After conditioning of the patient with cyclophosphamide, total
dysfunction, and diarrhea. Chronic GVHD, by definition, body irradiation, or other techniques to eradicate remaining
develops more than 100 days after transplantation. It is fre- malignant cells, the purged autologous stem cells are reinfused.
quently generalized in the form of a multisystem autoimmune Requirements for success in autologous transplantation are the
434 PART V Leukocyte Disorders

presence of normal pluripotent stem cells and reduction in the autologous transplants than among those receiving allogeneic
number of malignant cells to a level insufficient to cause recur- transplants.
rence from reinfused marrow. Even with the continued improvement in technique and sup-
A comparison of autologous transplantation with matched portive care, stem cell transplantation carries many risks. Death
allogeneic transplantation shows that (1) although allogeneic after transplantation is most likely caused by the following:
transplantation is not available to every patient, almost every 1. Complications of conditioning, such as infections or bleed-
patient is eligible for autologous transplantation; (2) among ing from bone marrow suppression
autologous transplant recipients, posttransplantation morbid- 2. Complications of GVHD
ity and mortality are lower and hospital stays are shorter; 3. Relapse (regrowth of malignant cells)
and (3) the relapse rate is higher among recipients of 4. Failure of donor cells to engraft

SUMMARY
Most malignancies of the hematopoietic system are acquired fashion, can lead to malignant transformation by disrupting the
genetic diseases. molecular machinery of the cell.
Most leukocyte malignancies are not localized, but rather are Most chromosomal translocations in leukemias involve onco-
systemic at the initiation of the malignant process. genes. The dominant transforming oncogene is able to alter the
For most leukocyte neoplasms, causes directly related to gene product and transform the cell into a malignant phenotype,
the development of the malignancy are unknown, but a few even in the presence of a residual normal allele.
exceptions exist. Some known causes include environmental In contrast to oncogenes, tumor suppressor genes contribute to
toxins, certain viruses, previous chemotherapy, and familial the malignant process only if both alleles have been lost or
predisposition. otherwise inactivated.
There are several classification schemes for leukocyte neoplasia, The formation of oncogenes or the loss of tumor suppressor genes
including the FAB system, based primarily on morphology and has molecular effects on the proliferation and differentiation of
cytochemical staining, and the WHO system, which retains some hematologic tissues.
elements of the FAB scheme but emphasizes molecular and cyto- Current treatments for leukocyte neoplasms can be roughly
genetic changes. divided into the following categories: chemotherapy, radiation
Chromosomal translocations in hematologic malignancies illus- therapy, supportive therapy, targeted therapy, and stem cell
trate that a single mutation, or a series of mutations in stepwise transplantation.

R E V I E W Q UESTIONS
1. Which of the following viruses is known to cause lym- 4. All of the following are among the cellular abnormalities
phoid malignancies in humans? produced by oncogenes except:
a. Human immunodeficiency virus a. Apoptotic failure
b. Epstein-Barr virus b. Suppression of DNA transcription
c. Hepatitis B virus c. Acceleration of DNA catabolism
d. Parvovirus d. Disruption of cell cycle processes

2. Tumor suppressor genes cause cancers such as leukemia 5. Chemotherapeutic agents are divided into which two
when mutations result in: major subgroups?
a. Suppression of cell division a. Phase specific and phase nonspecific
b. Failure to prevent malignant processes b. Toxic and nontoxic
c. Induction of cell division c. Oral and intravenous
d. Excessive apoptosis d. Traditional and modern

3. Oncogenes are said to act in a dominant fashion because: 6. G-CSF is provided as supportive treatment during leuke-
a. Cancer is a dominating disease that overtakes the mia treatment regimens to:
individuals health a. Suppress GVHD
b. The mutation product affects (i.e., dominates) normal b. Overcome anorexia
cells and mutated cells c. Prevent anemia
c. A mutation in a single allele is sufficient for d. Reduce the risk of infection
malignancy to develop
d. They are inherited by autosomal dominant
transmission
CHAPTER 29 Introduction to Leukocyte Neoplasms 435

7. Imatinib is an example of what type of leukemia 9. Hematopoietic stem cells for transplantation are harvested
treatment? from all of the following tissues except:
a. Supportive care a. Bone marrow
b. Chemotherapy b. Peripheral blood
c. Bone marrow conditioning agent c. Spleen
d. Targeted therapy d. Umbilical cord blood

8. Monoclonal antibodies may kill cancer cells by all of the 10. Compared with autologous bone marrow transplantation,
following mechanisms except: allogeneic transplantation has:
a. Binding to the cancer cells and causing the immune a. Better long-term success (i.e., no relapse)
system to destroy them b. Reduced occurrence of GVHD
b. Stimulating the production of anticancer cell c. Lower mortality rate
antibodies by the patients own immune system d. Better-tolerated conditioning phase
c. Activating complement on the cell surface
d. Increasing the rate of apoptosis in the cancer cells

REFERENCES
1. Jaffe ES, Harris NL, Stein H, et al, editors: In World Health 7. Westbrook CA: The molecular basis of neoplasia. In Hoffman
Organization classification of tumours: pathology and genetics of R, Benz EJ, Shattil SJ, et al, editors: Hematology: basic principles
tumours of haematopoietic and lymphoid tissues, Lyon, France, and practice, ed 4, Philadelphia, 2005, Churchill Livingstone,
2001, IARC Press. pp 941-954.
2. Swerdlow SH, Campo E, Harris NL, et al, editors: WHO clas- 8. Huntly B, Gilliland DG: Pathobiology of acute myeloid
sification of tumours of hematopoietic and lymphoid tissues, Lyon, leukemia. In Hoffman R, Benz EJ, Shattil SJ, et al, editors:
France, 2008, IARC Press. Hematology: basic principles and practice, ed 4, Philadelphia,
3. Rowley JD: A new consistent chromosomal abnormality in 2005, Churchill Livingstone, pp 1057-1069.
chronic myelogenous leukemia identified by quinacrine fluo- 9. Gaidano G, Dalla-Favera R: Pathobiology of non-Hodgkins
rescence and Giemsa staining. Nature 243:290-293, 1973. lymphomas. In Hoffman R, Benz EJ, Shattil SJ, et al, editors:
4. Taub R, Kirsch I, Morton C, et al: Translocation of the c-myc Hematology: basic principles and practice, ed 4, Philadelphia,
gene into the immunoglobulin heavy chain locus in human 2005, Churchill Livingstone, pp 1307-1324.
Burkitt lymphoma and murine plasmacytoma cells. Proc Natl 10. Stewart SJ: Immunotherapy. In Greer JP, Foerster J,
Acad Sci U S A 79:7837-7841, 1982. Lukens JN, et al, editors: Wintrobes clinical hematology,
5. Knudson AG: Hereditary cancer, oncogenes, and antionco- ed 11, Philadelphia, 2004, Lippincott Williams & Wilkins,
genes. Cancer Res 45:1437-1443, 1985. pp 2157-2177.
6. Dewald GW, Ketterling RP: Conventional cytogenetics 11. Kurzrock R, Kantarjian HM, Druker BJ, et al: Philadelphia
and molecular cytogenetics in hematologic malignancies. In chromosome-positive leukemias: from basic mechanisms
Hoffman R, Benz EJ, Shattil SJ, et al, editors: Hematology: basic to molecular therapeutics. Ann Intern Med 138:819-830,
principles and practice, ed 4, Philadelphia, 2005, Saunders, 2003.
pp 928-939.
30 Cytochemistry
Bernadette F. Rodak*

OUTLINE OBJECTIVES
Introduction and Principle After completion of this chapter, the reader will be able to:
Acceptable Specimens
and Fixatives 1. Discuss the purpose of performing cytochemical phosphatase, and tartrate-resistant leukocyte acid
Stains and Interpretations stains. phosphatase.
Myeloperoxidase 2. Determine appropriate specimen types, handling pro 4. Name the cell stage that is evaluated using the stains
Sudan Black B cedures, and fixatives for cytochemical stains, con listed in item 3.
Esterases sidering length of stability of specimens if staining is 5. Interpret the results of the stains listed in item 3.
Periodic AcidSchiff delayed. 6. Discuss the selection of controls in cytochemical
Factor VIII Antibodies 3. Discuss the principles and cell staining patterns for staining.
Leukocyte Alkaline the following tests: myeloperoxidase, Sudan black 7. Describe routine troubleshooting in cytochemical
Phosphatase B, esterases, periodic acidSchiff, leukocyte alkaline techniques.
Acid Phosphatase
(Tartrate Resistant)
Controls and
Troubleshooting

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 38-year-old woman came to the physician with a 2-month Sudan black B: negative
history of gingival bleeding and gingival hypertrophy. The -Naphthyl butyrate esterase: positive
CBC revealed the following: WBCs, 64 109/L; Hb, 7.5g/dL; Periodic acidSchiff: negative
and platelets, 36 109/L. The peripheral blood film and the 1. What cell lines stain positive for esterases?
bone marrow specimen demonstrated greater than 80% 2. What general diagnosis is suggested by the CBC values?
abnormal mononuclear cells. The results of cytochemical 3. Based on morphology and cytochemical staining, what is
testing of these cells were as follows: the classification of this disorder?
Myeloperoxidase: negative

Immunophenotyping, high-resolution cytogenetic studies, and


INTRODUCTION AND PRINCIPLE
molecular genetic analysis have reduced the importance of
Cytochemistry is the study of the chemical constituents of cells. cytochemical testing. Microarray technology can quantify the
These elements may be enzymatic (e.g., peroxidase) or nonen- expression of thousands of genes in a single analysis.1,2 However,
zymatic (e.g., lipids and glycogen). The cellular morphology of cytochemical testing still has limited use in many laboratories,
peripheral blood and bone marrow often provides a provisional especially in equivocal cases or where more sophisticated
diagnosis, and since the early twentieth century, cytochemical technology is not available.3,4 (Leukemias are discussed in
staining of cells has provided a useful adjunct for differentia- Chapters 29, 36, and 37; cytogenetics, in Chapter 31; molecular
tion of hematopoietic diseases, especially acute leukemias. diagnostics, in Chapter 32; and flow cytometry, in Chapter 33.)

*The author would like to acknowledge Carol Bradford for her work on this chapter in previous editions.

436
CHAPTER 30 Cytochemistry 437
Interpretation
ACCEPTABLE SPECIMENS AND FIXATIVES
MPO is present in the primary granules of granulocytic cells,
Many specimen types are adequate for cytochemical studies. beginning at the promyelocyte stage and continuing through-
Smears and imprints made from bone marrow, lymph nodes, out maturation. Leukemic myeloblasts also are usually positive
spleen, or peripheral blood are preferred. In enzymatic tech- for MPO. In many cases of the AMLs (without maturation, with
niques, fresh smears are used whenever possible to ensure maturation, and promyelocytic leukemia), it has been found
optimal enzyme activity. Smears for nonenzymatic stains, such that more than 80% of the blasts show MPO activity. Auer rods
as periodic acidSchiff (PAS) or Sudan black B (SBB), may found in leukemic blasts and promyelocytes test strongly MPO
remain stable for months if stored at room temperature. positive. Because of their strong MPO positivity, many Auer
Certain elements may be inhibited during the fixation of rods that could not be seen with a Wright-Giemsa stain can be
smears and imprints; it is important to use the proper fixative seen with the MPO stain.
for the desired cytochemical stains. Fixatives containing alcohol Monocytes are MPO negative to weakly or diffusely positive.
(methanol or ethanol), acetone, formaldehyde, or a combina- In contrast, lymphoblasts and lymphoid cells are MPO nega-
tion of these are commonly used for most cytochemical tive; in patients with ALL, fewer than 3% of the blasts show
studies. peroxidase positivity.8-10
It is important that the reaction only in the blast cells be
used as the determining factor for the differentiation of acute
leukemias. This is true for MPO and for the other cytochemical
STAINS AND INTERPRETATIONS
stains used in determining cell lineage that are mentioned in
Myeloperoxidase this chapter. The fact that maturing granulocytes are MPO posi-
Myeloperoxidase (MPO) (Figures 30-1 and 30-2) is an enzyme tive is normal and has little or no diagnostic significance.
found in the primary granules of neutrophils, eosinophils, and,
to a certain extent, monocytes. Lymphocytes do not exhibit Sudan Black B
MPO activity. This stain is useful for differentiating the blasts SBB staining (Figure 30-3) is another useful technique for the
of acute myeloid leukemia (AML) from those of acute lympho- differentiation of AML from ALL. The staining pattern is quite
blastic leukemia (ALL). similar to that of MPO; SBB staining is possibly a little more
sensitive for the early myeloid cells.
Principle
When hydrogen peroxide is present, MPO oxidizes dye sub- Principle
strates, creating black to red-brown staining (depending on SBB stains lipids, such as sterols, neutral fats, and phospholip-
the substrate) at the site of the activity. Benzidine had been ids, because of the solubility of the dye in lipid particles. These
the substrate most often used, but because of its carcinogenic lipids are found in the primary and secondary granules of
properties, other substrates such as 3,3-diaminobenzidine neutrophils and in the lysosomal granules of monocytes.6,11
or p-phenylenediamine dihydrochloride and catechol are cur-
rently used.5-7 Interpretation
Granulocytes (neutrophils) show a positive reaction to SBB
from the myeloblast through the maturation series. The stain-
ing becomes more intense as the cell matures as a result of the

Figure 30-2 Strong positive reaction to myeloperoxidase stain in leuke


Figure 30-1 Positive reaction to myeloperoxidase stain in early myeloid mic promyelocytes from a patient with acute promyelocytic leukemia (bone
cells. Note Auer rod at arrow (bone marrow, 1000). marrow, 1000).
438 PART V Leukocyte Disorders

increase in the numbers of primary and secondary granules. ducing a brightly colored compound at the site of the enzy-
Monocytic cells can demonstrate negative to weakly positive matic activity.5,12
staining, showing diffuse activity. Lymphoid cells generally do
not stain. In ALL, fewer than 3% of the blast cells show a posi- Interpretation
tive reaction.8-10 Esterase stains can be used to distinguish acute leukemias that
are granulocytic from leukemias that are primarily of mono-
Esterases cytic origin. When naphthol AS-D chloroacetate is used as a
Esterase reactions are used to differentiate myeloblasts and substrate, the reaction is positive in the granulocytic cells and
neutrophilic granulocytes from cells of monocytic origin. Nine negative to weak in the monocytic cells (Figure 30-4). Chloro-
isoenzymes of esterases are present in leukocytes. Two substrate acetate esterase is present in the primary granules of neutro-
esters commonly used are -naphthyl acetate and -naphthyl phils. Leukemic myeloblasts generally show a positive reaction.
butyrate (both nonspecific). Naphthol AS-D chloroacetate Auer rods show positivity as well.
(specific) also may be used. Specific refers to the fact that -Naphthyl acetate, in contrast to naphthol AS-D chloro
only granulocytic cells show staining, whereas nonspecific acetate, reveals strong esterase activity in monocytes that can
stains may produce positive results in other cells as well. be inhibited with the addition of sodium fluoride.8,13 Granu-
locytes and lymphoid cells generally show a negative result on
Principle nonspecific esterase staining (Figure 30-5).
Esterases hydrolyze an ester. A naphthol compound is released A positive -naphthyl butyrate esterase reaction is also
and combines with a diazonium salt (generally, hexazotized seen in monocytes. -Naphthyl butyrate is less sensitive than
pararosaniline, hexazotized new fuchsin, or fast blue BB), pro- -naphthyl acetate, but is more specific. Granulocytes and

Figure 30-3 Sudan black B reaction. The positivity increases with the Figure 30-4 Positive reaction to AS-D chloroacetate esterase stain in two
maturity of the granulocytic cell (bone marrow, 1000). granulocytic cells (bone marrow, 1000).

A B
Figure 30-5 A, Positive reaction to -naphthyl acetate esterase stain in monocytes (bone marrow, 1000). B, Same specimen with addition of sodium
fluoride. The esterase reaction in the monocytes is inhibited (bone marrow, 1000).
CHAPTER 30 Cytochemistry 439

Figure 30-6 -Naphthyl butyrate esterase positivity in cells of monocytic Figure 30-7 Periodic acidSchiff reaction showing coarse (block)
origin from a patient with acute monoblastic/monocytic leukemia. Note the positivity in a specimen from a patient with acute lymphoblastic leukemia
negativity of myeloid and erythroid precursors (bone marrow, 1000). (bone marrow, 1000).

lymphoid cells generally show a negative reaction (Figure


30-6). In myelomonocytic leukemia, positive AS-D chloroac-
etate activity and positive -naphthyl butyrate or -naphthyl
acetate activity should be seen because myeloid and monocytic
cells are present. In myelomonocytic leukemia, at least 20%
of the cells must show monocytic differentiation that is
nonspecific esterase positive and is inhibited by sodium fluo-
ride. In the pure monocytic leukemias, 80% or more of the
blasts are nonspecific esterase positive and specific esterase
negative.

Periodic AcidSchiff
PAS staining may be helpful in the diagnosis of some ALLs and
the erythroid type of AML, as well as in identifying abnormal
erythroid precursors in myelodysplastic syndromes. Figure 30-8 Periodic acidSchiff reaction showing coarse granular posi
tivity around the nucleus of early erythroid precursors in a specimen from a
patient with acute erythroid leukemia (bone marrow, 1000).
Principle
Many different cell types contain glycogen. Periodic acid oxi-
dizes glycogen, mucoproteins, and other high-molecular- normoblasts, or diffuse, which is more commonly seen in late
weight carbohydrates to aldehydes. These aldehydes react with normoblasts (Figure 30-8).14,15
the colorless Schiff reagent, which results in a bright red-pink
staining. The intensity of the staining depends on the number Factor VIII Antibodies
of aldehyde groups liberated by the periodic acid. The PAS Megakaryoblastic leukemia requires immunocytochemical
stain pattern may be fine and diffuse, coarse and granular stains for an accurate diagnosis. Monoclonal or polyclonal
(block), or a mixture of both. antibodies against factor VIIIrelated antigen or platelet glyco-
protein have given positive results in megakaryoblastic leuke-
Interpretation mia (Figure 30-9).16 A simplified cytochemical reaction chart
Granulocytes stain positively with PAS; the intensity of staining intended for quick reference is shown in Table 30-1.
increases as the cell matures. Megakaryocytes exhibit finely
diffuse staining, whereas platelets turn intensely red-pink. Leukocyte Alkaline Phosphatase
Normal erythrocyte precursors do not stain. Leukocyte alkaline phosphatase (LAP) enzyme activity is useful
In ALL, cells show a varied staining pattern. Lymphoblasts for differentiating chronic myelogenous leukemia (CML) from
of ALL may show a coarse block pattern of activity, a finely a leukemoid reaction that may be seen in severe infections.
diffuse pattern, or a combination of the two patterns, or they
may show no staining (Figure 30-7). Cells from Burkitt lym- Principle
phoma generally do not stain.10,11 LAP is an enzyme found in the membranes of secondary gran-
In the erythroid type of AML and in myelodysplastic syn- ules of neutrophils. The substrate naphthol AS-BI phosphate is
dromes, PAS-positive erythroid precursors may be found. The hydrolyzed by the enzyme at an alkaline pH. This hydrolyzed
staining pattern may be coarse and granular, especially in early substrate, in combination with a dye such as fast red-violet LB
440 PART V Leukocyte Disorders

TABLE 30-1 Simplified Acute Leukemia Reaction Chart


CYTOCHEMICAL STAIN
Condition MPO SBB NASDA ANBE ANAE PAS Factor VIII
ALL /+ (focal) /+ (focal) Varied
AML + + + Varied
AMML + + + + (diffuse) + (diffuse) Varied
AMoL /+ + (diffuse) + (diffuse) Varied
Erythroleukemia * * * +; blocky in pronormoblasts
Megakaryocytic leukemia + (localized) /+ (localized) +

+, Positive reaction; , negative reaction; /+, positive or negative reaction; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; AMML, acute myelomonocytic
leukemia; AMoL, acute monocytic leukemia; ANAE, -naphthyl acetate esterase; ANBE, -naphthyl butyrate esterase; MPO, myeloperoxidase; NASDA, naphthol AS-D chloroacetate
esterase; PAS, periodic acidSchiff; SBB, Sudan black B.
*Positive reaction in myeloblasts; negative reaction in normoblasts.

TABLE 30-2 Results of Leukocyte Alkaline


Phosphatase Staining*
Condition Score
Normal 20-100
Chronic myelogenous leukemia <13
Leukemoid reaction >100
Polycythemia vera 100-200
Secondary polycythemia 20-100

*May vary slightly with ranges determined in each laboratory.

Interpretation
Figure 30-9 Positive reaction to factor VIII staining in a specimen from a A normal LAP score should be between 20 and 100, but
patient with acute megakaryocytic leukemia (bone marrow, 1000). because of scoring subjectivity, the range can extend as low as
15 or as high as 170. Thus, it is important that every laboratory
or fast blue BB, produces a colored precipitate at the site of the establish its own reference intervals. Specimens from individu-
enzymatic activity. als with untreated CML have decreased LAP scores; those from
individuals with leukemoid reactions have scores ranging from
Scoring high normal to increased. Other conditions associated with
LAP activity is scored in the mature segmented cells and bands higher scores are the third trimester of pregnancy and polycy-
only. The activity scores range from 0 to 4+ (Figure 30-10). The themia vera. The score is normal in cases of secondary polycy-
scores of 100 mature segmented cells and bands are added to themia. Other conditions in which decreased scores are found
determine the LAP score. For example: include paroxysmal nocturnal hemoglobinuria, sideroblastic
anemia, and myelodysplastic disorders. A summary of LAP
Score No. Cells Score No. Cells interpretation is given in Table 30-2.
0 20 0
1 45 45 Acid Phosphatase (Tartrate Resistant)
2 25 50 Almost all blood cells contain seven nonerythroid isoenzymes
3 5 15 of acid phosphatase: 0, 1, 2, 3, 3b, 4, and 5.17,18 Hairy cells
4 5 20 produce isoenzyme 5 in abundance. This makes the acid phos-
Total 100 130 = LAP score phatase stain a useful diagnostic tool for confirmation of hairy
cell leukemia.
Because scoring is subjective, it is recommended that two
slides be scored by two people. The LAP scores should differ Principle
by less than 10%. If the scores do not agree, a third slide from Acid phosphatase hydrolyzes the substrate naphthol AS-BI
the patient must be scored. phosphoric acid. When hydrolyzed, this substrate couples
Eosinophils do not show alkaline phosphatase activity and with a dye such as fast garnet GBC and produces a red pre
must not be mistaken for mature neutrophils with a score cipitate at the site of the enzymatic activity. When L-(+)-
of zero. Eosinophils can be distinguished by their larger tartaric acid is added, all isoenzymes except isoenzyme 5 are
granules. inhibited. Isoenzyme 5 is tartrate resistant. The term TRAP
CHAPTER 30 Cytochemistry 441

A B

C D

E
Figure 30-10 Specimens stained for leukocyte alkaline phosphatase showing activity scores from 0 to 4+ (peripheral blood, 1000). A, 0. B, 1+. C, 2+.
D, 3+. E, 4+.
442 PART V Leukocyte Disorders

TABLE 30-3 Results of Leukocyte Acid Phosphatase


Staining
Cell Type Without Tartrate With Tartrate
Lymphocytes +
Hairy cells + +

+, Positive reaction; , negative reaction.

hematopoietic cells contained in the marrow aspirate can serve


as an internal control for staining. For hairy cell leukemia, a
normal blood film can be used to test inhibition by L-(+)-
tartaric acid, but it is more difficult to obtain a control for
Figure 30-11 Positive acid phosphatase reaction. If hairy cells are tartrate resistance. Only a blood film from a patient known to
present, the stain results remain positive after the addition of tartaric acid to have hairy cell leukemia can be used.
the incubation mixture (peripheral blood, 1000).
When the control slides do not exhibit the proper staining
pattern, such as showing no activity when they should be posi-
tive or exhibiting the wrong color of precipitate, the investiga-
(tartrate-resistant acid phosphatase) staining is often used to tion should include the possibility that a wrong reagent, no
denote this phenomenon. reagent, or an expired reagent was used in the test system. A
reagent may have been contaminated; if this is the case, the
Interpretation examiner should make or open a new batch of reagent. If the
Most hematopoietic cells show acid phosphatase activity. reagent is acceptable, the examiner should go over the proce-
Granulocytes, lymphocytes, and monocytes all are positive to dure to ensure that all steps were followed correctly. Another
some extent for this activity until L-(+)-tartaric acid is added. aspect to consider is how old the film is and how it was stored.
The activity is inhibited because isoenzyme 5 is lacking. Hairy Some enzymes diminish in activity over time. Fresh films
cells, which contain isoenzyme 5, remain positive with the are always best. Films with the -naphthyl acetate esterase,
addition of L-(+)-tartaric acid. In hairy cell leukemia, at least -naphthyl acetate esterase with sodium fluoride, and TRAP
two or more cells contain 40 or more red granules (Figure stains should not be coverslipped with routine mounting
30-11, Table 30-3).5 media, because the stain fades. However, a neutral mounting
medium can be used, or stained films can be viewed directly
under oil immersion with no effect on reaction products.8
CONTROLS AND TROUBLESHOOTING
If the control results are unacceptable, the cytochemical
For most cytochemical stains mentioned in this chapter, a stain in question must be repeated. Specific directions for test
normal blood film containing neutrophils, lymphocytes, and performance are included in test kits or can be found in an
monocytes is sufficient as a control sample. A few normal atlas of cytochemistry.5,19

SUMMARY
Cytochemical stains aid in the differentiation of morphologically Esterases help differentiate granulocytes and their precursors
similar cells by identifying enzymes, lipids, glycogen, or other from cells of monocytic origin. Butyrate esterase testing gives
substances in cells. positive results in monocytes but not in granulocyte precursors,
Cytochemical techniques are often used in conjunction with mor- whereas naphthol AS-D chloroacetate esterase stains granulocyte
phologic analysis, immunohistochemical methods, flow cytometry, precursors.
cytogenetic analysis, and molecular biologic techniques in estab- PAS stains yield positive results in specimens from patients with
lishing a diagnosis. some ALLs and with erythroid precursors in erythroid leukemia
Cytochemical reactions may be enzymatic or nonenzymatic. Fresh and myelodysplastic syndromes.
smears must be used to detect enzymatic activity, whereas non- Megakaryocytic leukemia requires an immunohistochemical stain
enzymatic procedures may be performed on specimens that have for antibodies against factor VIIIrelated antigen or platelet
been stored at room temperature. glycoprotein.
MPO stains primary granules and is useful in differentiating granu- LAP is most useful in distinguishing CML from leukemoid reactions.
locytic from lymphoid cells. Specimens from patients with CML have a very low LAP score.
SBB stains lipids and results parallel those with the MPO Acid phosphatase with tartrate inhibition yields positive results in
stain. specimens from patients with hairy cell leukemia.
CHAPTER 30 Cytochemistry 443

For most cytochemical stains mentioned in this chapter, a normal Now that you have completed this chapter, go back and
blood film containing neutrophils, lymphocytes, and monocytes is read again the case study at the beginning and respond
sufficient as a control sample. If results for the control samples to the questions presented.
are unacceptable, the cytochemical stain in question must be
repeated.

R E V I E W Q UESTIONS
1. Smears for application of which of the following cyto- 7. Which of the following stains is used to evaluate mature
chemical stains may be stable for months if stored at room neutrophils and bands?
temperature? a. Acid phosphatase
a. MPO b. LAP
b. SBB c. PAS
c. LAP d. SBB
d. -Naphthyl butyrate esterase
8. Cytochemical staining was performed on the cells from a
2. Which cytochemical stain can be used to differentiate AML spleen imprint for a patient with suspected leukemia. The
from ALL? staining was performed promptly, and the infiltrating cells
a. TRAP stained positive for acid phosphatase. The acid phospha-
b. LAP tase staining was repeated on a second imprint after the
c. MPO slide was pretreated with tartrate. The specimen still tested
d. -Naphthyl acetate positive for acid phosphatase. This result suggests which
of the following conditions?
3. SBB stains which of the following component of cells? a. Erythrocytic leukemia
a. Glycogen b. AML
b. Lipids c. Hairy cell leukemia
c. Structural proteins d. Acute megakaryoblastic leukemia
d. Enzymes
9. A normal blood film provides a suitable positive control
4. Hairy cells produce an abundance of which isoenzyme of for testing with all of the following stains except:
acid phosphatase? a. MPO
a. 1 b. -Naphthyl acetate esterase
b. 3b c. Acid phosphatase with tartrate pretreatment
c. 4 d. LAP
d. 5
10. Which of the following is not a suitable fixative for slides
5. An LAP score of 250 would be consistent with a diagnosis that are to be cytochemically stained?
of: a. Phosphate buffered saline
a. Normal cells b. Methanol
b. CML c. Acetone
c. Hairy cell leukemia d. Formaldehyde
d. Leukemoid reaction

6. The cytochemical stain -naphthyl butyrate is a nonspe-


cific esterase stain that reacts with cells of which lineage?
a. Erythroid
b. Monocytic
c. Granulocytic
d. Lymphoid

REFERENCES
1. Haferlach T, Kohlmann A, Bacher U, et al: Gene expression 2. Basso G, Case C, DellOrto MC: Diagnosis and genetic sub-
profiling for the diagnosis of acute leukemia. Br J Cancer types of leukemia combining gene expression and flow
96:535-540, 2007. cytometry. Blood Cells Mol Dis 39:164-168, 2007.
444 PART V Leukocyte Disorders

3. Bain BJ: Routine and specialized techniques in the diagnosis 12. Li C-Y, Lam KW, Yam LT: Esterases in human leukocytes.
of haematological neoplasms. J Clin Pathol 48:501-508, 1995. J Histochem Cytochem 21:1-12, 1973.
4. Orazi A: Histopathology in the diagnosis and classification 13. Wachstein M, Wolf G: The histochemical demonstration of
of acute myeloid leukemia, myelodysplastic syndromes, esterase activity in human blood and bone marrow smears.
and myelodysplastic/myeloproliferative diseases. Pathobiology J Histochem Cytochem 6:457, 1958.
74:97-114, 2007. 14. Davey FR, Abraham N, Brunetto VL, et al: Morphologic
5. Li C-Y, Yam LT, Sun T: Modern modalities for the diagnosis characteristics of erythroleukemia (acute myeloid leukemia;
of hematologic neoplasms: color atlas/text, New York, 1996, FAB-M6): a CALGB study. Am J Hematol 49:29-38, 1995.
Igaku-Shoin. 15. Quaglino D, Hayhoe FGJ: Periodic-acidSchiff positivity in
6. Li C-Y, Yam LT: Cytochemistry and immunochemistry in erythroblasts with special reference to Di Guglielmos disease.
hematologic diagnoses. Hematol Oncol Clin North Am 8:665- Br J Haematol 6:26-33, 1960.
681, 1994. 16. Bennett JM, Catovsky D, Daniel MT, et al: Criteria for the
7. Sheibani K, Lucas FV, Tubbs RR, et al: Alternate chromogens diagnosis of acute leukemia of megakaryocyte lineage (M7).
as substitutes for benzidine for myeloperoxidase cytochem- Ann Intern Med 103:460-462, 1985.
istry. Am J Clin Pathol 75:367-370, 1981. 17. Hoyer JD, Li C-Y, Yam LT, et al: Immunohistochemical
8. Scott CS, Den Ottolander GJ, Swirsky D, et al: Recommended demonstration of acid phosphatase isoenzyme 5 (tartrate-
procedures for the classification of acute leukemias. Leuk Lym- resistant) in paraffin sections of hairy cell leukemia and
phoma 11:37-50, 1993. other hematologic disorders. Am J Clin Pathol 108:308-315,
9. Cheson BD, Cassileth DR, Schiffer D, et al: Report of the 1997.
National Cancer Institutesponsored workshop on defini- 18. Li C-Y, Yam LT, Lam KW: Acid phosphatase isoenzyme in
tions of diagnosis and response in acute myeloid leukemia. human leukocytes in normal and pathologic conditions.
J Clin Oncol 8:813-819, 1990. J Histochem Cytochem 18:473-481, 1970.
10. Head DR: Revised classification of acute myeloid leukemia. 19. Sun T, Li C-Y, Yam LT: Atlas of cytochemisty and immunocyto-
Leukemia 10:1826-1831, 1996. chemisty of hematologic neoplasms, Chicago, 1985, American
11. Hayhoe FJG: The cytochemical demonstration of lipids in Society of Clinical Pathologists Press.
blood and bone marrow cells. J Pathol Bacteriol 65:413-421,
1953.

ADDITIONAL RESOURCES
Swerdlow SH, Campo E, Harris NL, et al, editors: WHO classifica-
tion of tumours of haematopoietic and lymphoid tissues, ed 4, Lyon,
2008, IARC Press.
Cytogenetics
Gail H. Vance
31
OUTLINE OBJECTIVES
Reasons for Chromosome After completion of this chapter, the reader will be able to:
Analysis
Chromosome Structure 1. Describe chromosome structure and the methods 7. Describe the types of chromosomal abnormalities
Cell Cycle used in G-banded chromosome identification. that are detectable with cytogenetic methods.
Chromosome Architecture 2. Explain the basic laboratory techniques for preparing 8. Given the designation of a chromosome mutation, be
Metaphase Chromosomes chromosomes for analysis. able to determine whether the abnormality is numeric
Chromosome 3. Differentiate between numeric and structural chro- or structural, which chromosomes are affected, what
Identification mosome abnormalities. type of abnormality it is, and what portion of the
Chromosome Number 4. Discuss the importance of karyotype in the diagnosis chromosome is affected.
Chromosome Size and of hematologic cancer. 9. Given a diagram of a G-banded generic chro
Type 5. Explain the basic technique of fluorescence in situ mosome, name the structures identifiable by light
Techniques for
hybridization (FISH). microscopy.
Chromosome
6. Discuss the advantage of using FISH analysis in 10. Define array comparative genomic hybridization.
Preparation and
Analysis conjunction with G-banded analysis of cells.
Chromosome Preparation
Chromosome Banding CASE STUDY
Metaphase Analysis
Nonradioactive in Situ After studying the material in this chapter, the reader should be able to respond to the following
Hybridization case study:
Cytogenetic
A 54-year-old man came to his physician with a history of fatigue, weight loss, and increased bruis-
Nomenclature
Chromosome ing over a 6-month period. His WBC count was elevated at 200 109/L. A bone marrow aspirate
Abnormalities was sent for cytogenetic analysis. G-banded chromosome analysis of 20 cells from bone marrow
Numeric Abnormalities cultures showed all cells to be positive for the Philadelphia chromosome, t(9;22)(q34;q11.2), as
Structural Aberrations seen in chronic myelogenous leukemia (Figure 31-1). FISH studies using the BCR and ABL gene
Cancer Cytogenetics
Leukemia
Solid Tumors
Array Comparative
Genomic Hybridization

Figure 31-1 Karyogram for the patient in the case study showing a translocation between chromosomes 9 and 22,
characteristic of chronic myelogenous leukemia. (Courtesy the Cytogenetics Laboratory, Indiana University School of
Medicine, Indianapolis, Ind.)
Continued

445
446 PART V Leukocyte Disorders

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

probes (Abbott Molecular, Des Plaines, Ill.) produced dual


BCR/ABL1
fusion signals, one located on the derivative chromosome 9
and one on the derivative chromosome 22, characteristic of
the translocation between chromosomes 9 and 22 leading to
the rearrangement of BCR and ABL1 oncogenes (Figure 31-2).
The patient was treated with imatinib mesylate for the next
2 months. Another cytogenetic study was performed on a
second bone marrow aspirate. This analysis showed that 12
of 20 cells analyzed were normal, 46,XY[12]; however, there
were still 8 cells positive for the Philadelphia chromosome,
46,XY,t(9;22)(q34; q11.2)[8].
1. What is G-banded chromosome analysis?
2. Is the described mutation an example of a numeric or a
structural abnormality? What type? Which chromosomes
are involved? Explain.
3. What is FISH and how does it complement standard chro-
mosome analysis?

Figure 31-2 Bone marrow metaphase cell for the patient in the case
study hybridized with probes for BCR (green) and ABL (red) (Abbott
Molecular, Des Plaines, Ill.). The fusion signals (yellow) represent the
translocated chromosomes 9 and 22. (Courtesy the Cytogenetics Labora-
tory, Indiana University School of Medicine, Indianapolis, Ind.)

H
uman cytogenetics is the study of chromosomes, nonrandom chromosome abnormalities are recognized in
their structure, and their inheritance. There are many forms of cancer.
approximately 25,000 genes in the human genome, Physicians who care for patients of all ages may order chro-
most of which reside on the 46 chromosomes normally found mosome analysis or karyotyping for patients with mental retar-
in each somatic cell.1 dation, infertility, ambiguous genitalia, short stature, fetal loss,
Chromosome disorders are classified as structural or numer- risk of genetic or chromosomal disease, and cancer. In the fol-
ical and involve the loss, gain, or rearrangement of either a lowing discussion, basic cytogenetic concepts are presented.
piece of a chromosome or the entire chromosome. Because This field is in a period of tremendous growth. Supplementation
each chromosome contains thousands of genes, a chromo- of this chapter with the material in Chapter 32 is recommended.
somal abnormality that is observable by light microscopy
involves, on average, 3 to 5 megabases of DNA and represents
CHROMOSOME STRUCTURE
the disruption or loss of hundreds of genes. Such disruptions
often have a profound clinical effect. Chromosomal abnor- Cell Cycle
malities are observed in approximately 0.65% of all live births.2 The cell cycle is divided into four stages: G1, the growth period
The gain or loss of an entire chromosome, other than a sex before synthesis of deoxyribonucleic acid (DNA); S phase, the
chromosome, is usually incompatible with life and accounts period during which DNA synthesis takes place; G2, the period
for approximately 50% of first-trimester spontaneous abor- after DNA synthesis; and M, the period of mitosis or cell divi-
tions.3 In leukemia, cytogenetic abnormalities are observed in sion, the shortest phase of the cell cycle (Figure 31-3). During
more than 50% of bone marrow specimens.4 These recurring mitosis, chromosomes are maximally condensed. While in
abnormalities often define the leukemia and frequently indi- mitosis, cells can be chemically treated to arrest cell progres-
cate clinical prognosis. sion through the cycle so that the chromosomes can be isolated
and analyzed.

REASONS FOR CHROMOSOME ANALYSIS


Chromosome Architecture
Chromosome analysis is an important diagnostic procedure in A chromosome is formed from a single long DNA molecule
clinical medicine. Not only are chromosomal anomalies major that contains a series of genes. The complementary double-
causes of reproductive loss and birth defects, but also helix structure of DNA was established in 1953 by Watson and
CHAPTER 31 Cytogenetics 447

Figure 31-3 Cell cycle.

Crick.5 The backbone is a sugar-phosphate-sugar polymer. The


sugar is deoxyribose. Attached to the backbone and filling the
center of the helix are four nitrogen-containing bases. Two of
these, adenine (A) and guanine (G), are purines; the other two,
cytosine (C) and thymine (T), are pyrimidines (Figure 31-4).
The chromosomal DNA of the cell resides in the cells
nucleus. This DNA and its accompanying proteins are referred
to as chromatin. During the cell cycle, at mitosis, the nuclear
chromatin condenses approximately 10,000-fold to form chro-
mosomes.6 Each chromosome results from progressive folding
and compaction of the entire nuclear chromatin. This conden-
sation is achieved through multiple levels of helical coiling and
supercoiling (Figure 31-5).

Metaphase Chromosomes
Metaphase is the stage of mitosis where the chromosomes align
on the equatorial plate. Electron micrographs of metaphase
chromosomes have provided models of chromosome struc- Figure 31-4 The two DNA chains are held together by the pairing of bases
ture. In the beads-on-a-string model of chromatin folding, on the interior of the helix. Hydrogen bonds unite a purine base with a
pyrimidine base. A, Adenine; C, cytosine; G, guanine; T, thymine. (From
the DNA helix is looped around a core of histone proteins.7
Watson JD, Tooze J, Kurtz DT: Recombinant DNA: a short course, New York,
This packaging unit is known as a nucleosome and measures
1983, WH Freeman, p 19. 1983 James D. Watson, John Tooze, and
approximately 11 nm in diameter.8 Nucleosomes are coiled David T. Kurtz. Used with the permission of WH Freeman and Company.)
into twisted forms to create an approximately 30-nm chroma-
tin fiber. This fiber, called a solenoid, is condensed further and
bent into a loop configuration. These loops extend at an angle chromosomes. The reindeer has a relatively high chromosome
from the main chromosome axis.9 number for a mammal (2n = 76), whereas the Indian muntjac,
or barking deer, has a very low chromosome number (2n = 7
in the male and 2n = 6 in the female).11
CHROMOSOME IDENTIFICATION
Chromosome Number Chromosome Size and Type
In 1956, Tijo and Levan10 identified the correct number of In the 1960s, before the discovery of banding, chromosomes
human chromosomes as 46. This is the diploid chromosome were categorized by overall size and the location of the centro-
number and is determined by counting the chromosomes in mere (primary constriction) and were assigned to one of seven
dividing somatic cells. The designation for the diploid number groups, A through G. Group A includes chromosome pairs 1,
is 2n. Gametes (ova and sperm) have half the diploid number 2, and 3. These are the largest chromosomes, and their centro-
(23). This is called the haploid number of chromosomes and is meres are located in the middle of the chromosome (i.e., they
designated as n. Different species have different numbers of are metacentric). Group B chromosomes, pairs 4 and 5, are the
448 PART V Leukocyte Disorders

Chromatid
Telomere

Satellite
Stalk

Centromere

Submetacentric Acrocentric
Chromosome
metacentric
Figure 31-6 Chromosome morphology. The three shapes of chromo-
somes are metacentric, submetacentric, and acrocentric. This figure also
shows the position of the centromere and telomere as well as the two chro-
matids that comprise a chromosome.

Phytohemagglutinin primarily stimulates T cells to divide,12


whereas pokeweed preferentially stimulates B lymphocytes.13
Chromosomes can be obtained from replicating cells by
arresting the cell in metaphase. Cells from the peripheral blood
or bone marrow are cultured in media for 24 to 72 hours. In
standard peripheral blood cultures, a mitogen is added to
stimulate cellular division. Neoplastic cells are spontaneously
dividing and generally do not require stimulation with a
mitogen. After the cell cultures have grown for the appropriate
period, Colcemid, an analogue of colchicine, is added to
disrupt the mitotic spindle fiber attachment to the chromo-
some. After culture, cells are exposed to a hypotonic (potas-
Figure 31-5 Chromosome structure. The folding and twisting of the DNA sium chloride) solution that causes the chromosomes to spread
double helix. (From Gelehrter TD, Collins FS, Ginsberg D: Principles of apart from one another. A fixative of methanol and acetic acid
medical genetics, ed 2, Philadelphia, 1998, Lippincott Williams & Wilkins.) is added that hardens cells and removes proteinaceous mate-
rial. Cells are dropped onto cold, wet glass slides to achieve
optimal dispersal of the chromosomes. The slides are then aged
next largest chromosomes; their centromeres are off center, or before banding.
submetacentric. Group G consists of the smallest chromo-
somes, pairs 21 and 22, whose centromeres are located at the Chromosome Banding
ends of the chromosomes and are designated as acrocentric Analysis of each chromosome is made possible by staining
(Figure 31-6). with a dye. The name chromosome is derived from the Greek
words chroma, meaning color, and soma, meaning body.
Hence chromosome means colored body. In 1969, Caspers-
TECHNIQUES FOR CHROMOSOME
son, Zech, Modest, etal14 were the first investigators to stain
PREPARATION AND ANALYSIS
chromosomes successfully with a fluorochrome dye. Using
Chromosome Preparation quinacrine mustard, which binds to adenine-thyminerich
Tissues used for chromosome analysis contain cells with an areas of the chromosome, they were able to distinguish a
inherently high mitotic rate (bone marrow cells) or cells banding pattern unique to each chromosome. This banding
that can be stimulated to divide in culture (peripheral blood pattern, called Q banding, differentiates the chromosome into
lymphocytes). Special harvesting procedures are established bands of differing widths and relative brightnesses (Figure
for each tissue. Mitogens such as phytohemagglutinin or 31-7). The most brightly fluorescent bands of the 46 human
pokeweed mitogen are added to peripheral blood cultures. chromosomes include the distal end of the Y chromosome, the
CHAPTER 31 Cytogenetics 449

Figure 31-7 Q-banded preparation. Note the intense brilliance of Yq. (Courtesy the Cytogenetics Laboratory, Indiana University School of Medicine,
Indianapolis, Ind.)

centromeric regions of chromosomes 3 and 4, and the short


arms of the acrocentric chromosomes (13, 14, 15, 21, and 22).
Other stains are used to identify chromosomes, but in con-
trast to Q banding, these methods normally necessitate some
pretreatment of the slide to be analyzed. Giemsa (G) bands are
obtained by pretreating the chromosomes with the proteolytic
enzyme trypsin. GTG banding means G banding by Giemsa
with the use of trypsin. Giemsa, like quinacrine mustard,
stains AT-rich areas of the chromosome. The dark bands are
called G-positive (+) bands. Guanine-cytosinerich areas of the
chromosome have little affinity for the dye and are referred to
as G-negative () bands. G+ bands correspond with the brightly
fluorescing bands of Q banding (Figures 31-8 and 31-9). G
banding is the most common method used for staining
chromosomes.
C banding stains the centromere (primary constriction) of
Figure 31-8 Normal male metaphase chromosomes.
the chromosome and the surrounding condensed heterochro-
matin. Constitutive heterochromatin is a special type of late-
replicating repetitive DNA that is located primarily at the (NORs). NORs contain tandemly repeated ribosomal ribonu-
centromere of the chromosome. In C banding, the chromo- cleic acid (RNA) genes. NORs can be differentially stained in
somes are treated first with an acid and then with an alkali chromosomes by a silver stain in a method called AG-NOR
(barium hydroxide) before Giemsa staining. C banding is most banding.
intense in human chromosomes 1, 9, and 16 and the Y chro- Chromosome banding is visible after chromosome conden-
mosome. Polymorphisms are also observed in the C bands sation, which occurs during mitosis. The banding pattern
from different individuals. These polymorphisms have no observed depends on the degree of condensation. By examina-
clinical significance (Figure 31-10). tion of human chromosomes early in mitosis, it has been
Specific chromosomal regions that are associated with the possible to estimate a total haploid genome (23 chromosomes)
nucleoli in interphase cells are called nucleolar organizer regions with approximately 2000 AT-rich (G+) bands.15 The later the
450 PART V Leukocyte Disorders

noted, the technologist may need to analyze additional cells.


Computer imaging or photography is used to confirm and
record the microscopic analysis. A picture of all the chromo-
somes aligned from 1 to 22 including the sex chromosomes is
called a karyogram.

Nonradioactive in Situ Hybridization


The use of molecular methods coupled with standard karyo-
type analysis has improved chromosomal detection capability
beyond that of the light microscope. DNA or RNA probes
labeled with either fluorescent or enzymatic detection systems
are hybridized directly to metaphase or interphase cells on a
glass microscope slide. These probes usually belong to one of
three classes: (1) probes for repetitive DNA sequences, primar-
ily generated from centromeric DNA; (2) whole-chromosome
probes that include segments of an entire chromosome; and
Figure 31-9 Normal male karyogram, GTG-banded preparations.
(Courtesy the Cytogenetics Laboratory, Indiana University School of Medicine, (3) specific loci or single-copy probes.
Indianapolis, Ind.) Fluorescence in situ hybridization (FISH) is a molecular
technique commonly used in cytogenetic laboratories. FISH
studies are a valuable adjunct to the diagnostic workup. In
FISH, the DNA or RNA probe is labeled with a fluorophore.
Target DNA is treated with heat and formamide to denature
the double-stranded DNA, which renders it single-stranded.
The target DNA anneals to a similarly denatured, single-
stranded, fluorescently labeled DNA or RNA probe with a
complementary sequence. After hybridization, the unbound
probe is removed through a series of stringent washes, and the
cells are counterstained for visualization (Figure 31-11).
In situ hybridization with centromere or whole-chromosome
painting probes can be used to identify individual chromo-
somes (Figure 31-12). Marker chromosomes represent chroma-
tin material that has been structurally altered and cannot be
identified by a G-band pattern. FISH using a centromere or
paint probe, or both, is often helpful in identifying the chro-
mosome of origin (Figure 31-13).16 Specific loci probes can be
used to detect both structural and numerical abnormalities but
Figure 31-10 C-banded male metaphase chromosomes. (Courtesy the
Cytogenetics Laboratory, Indiana University School of Medicine, Indianapolis, are especially helpful in identifying chromosomal transloca-
Ind.) tions or inversions.
The FISH procedure has many advantages and has advanced
the detection of chromosomal abnormalities beyond that of
stage of mitosis, the more condensed the chromosome and the G-banded analysis. Both dividing (metaphase) and nondivid-
fewer total G+ bands observed. ing (interphase) cells can be analyzed with FISH. Performance
of FISH on uncultured cells, such as bone marrow smears,
Metaphase Analysis provides a quick test result that can be reported in 24 hours.
After banding, prepared slides with dividing cells are scanned Also, in some cultured bone marrow samples, the number of
under a light microscope with a low-power objective lens dividing cells may be insufficient for cytogenetic diagnosis by
(10). When a metaphase cell has been selected for analysis, a G-banded metaphase analysis. In such cases, FISH performed
63 or 100 oil immersion objective lens is used. Each meta- on interphase (nondividing) cells with probes for a specific
phase cell is analyzed first for chromosome number. Then each translocation may provide the diagnosis. FISH also can be
chromosome pair is analyzed for its banding pattern. A normal performed on paraffin-embedded tissue sections, specimens
somatic cell contains 46 chromosomes, which includes 2 sex obtained by fine needle aspiration, and touch preparations
chromosomes and 22 pairs of autosomes (chromosomes 1 from lymph nodes or solid tumors.
through 22). The technologist records his or her summary of
the analysis using chromosome nomenclature. This summary
CYTOGENETIC NOMENCLATURE
is called a karyotype. Any variation in number and banding
pattern is recorded by the technologist. At least 20 metaphase Banding techniques enabled scientists to identify each chromo-
cells are analyzed from leukocyte cultures. If abnormalities are some pair by a characteristic banding pattern. In 1971, a Paris
CHAPTER 31 Cytogenetics 451

General FISH Protocol


Slide preparation
with interphase or
metaphase cells

Labeling Place probe on slide


DNA Probe Labeled Probe
Nick Translation Cover with coverslip
PCR

Denature 75 C

Detection

Apply antibodies Hybridize 37 C


overnight
Examine under Wash excess probe
fluorescent microscope
Figure 31-11 General protocol for fluorescence in situ hybridization. PCR, Polymerase chain reaction.

conference for nomenclature of human chromosomes was


convened to designate a system to describe the regions and
specific bands of the chromosomes. The chromosome arms
were designated p (petite) for the short arm and q for the long
arm. The regions in each arm and the bands contained within
each region were numbered consecutively, from the centro-
mere outward to the telomere or end of the chromosome. To
designate a specific region of the chromosome, the chromo-
some number is written first, followed by the designation of
either the short or long arm, then the region of the arm, and
finally the specific band. Xq21 designates the long arm of
the X chromosome, region 2, band 1. To designate a sub-
band, a decimal point is placed after the band designation,
followed by the number assigned to the sub-band, as in Xq21.1
(Figure 31-14).
Cytogenetic (and FISH) nomenclature represents a uniform
Figure 31-12 Metaphase preparation is painted with multiple probes code used by cytogeneticists to communicate chromosome
for chromosome 7, producing a fluorescent signal. (Courtesy the Cytogenet- abnormalities. In this nomenclature each string begins with
ics Laboratory, Indiana University School of Medicine, Indianapolis, Ind.) the modal number of chromosomes followed by the sex chro-
mosome designation. A normal male karyotype is designated
46,XY and a normal female karyotype is designated 46,XX.
Locus-specific probes If abnormalities are observed in the cell, the designation
Chromosome paint probes is written to include abnormalities of modal chromosome
Centromere probes number, sex chromosomes, and then the autosomes. A cell
with trisomy of chromosome 8 (three copies of chromosome
Detection of numerical Detection of structural
chromosome abnormalities chromosome abnormalities 8) in a male is written as 47,XY,+8. The number of cells with
this abnormality is indicated in brackets. If 20 cells were exam-
Trisomy Deletions ined, trisomy 8 was found in 10 cells, and the remainder
Monosomy Duplications were normal, the findings would be written as 47,XY,+8[10]/
Polyploidy Inversions
Translocations 46,XY[10]. Translocations (exchange of material between two
Ring chromosomes chromosomes) are designated t, with the lowest chromosome
Marker chromosomes number listed first. Thus a translocation between the short
Dicentric chromosomes arm of chromosome 12 at band p13 and the long arm of
Figure 31-13 Fluorescence in situ hybridization in the clinical laboratory. chromosome 21 at band q22 is written as t(12;21)(p13;q22).
452 PART V Leukocyte Disorders

1 2 3 4 5

6 7 8 9 10 11 12

13 14 15 16 17 18

19 20 21 22 X Y
Figure 31-15 Triploid karyotype, 69,XXY. (Courtesy the Cytogenetics
Laboratory, Indiana University School of Medicine, Indianapolis, Ind.)

Figure 31-14 Banding pattern of the human X chromosome at the 550-


band level.

A semicolon is used to separate the chromosomes. A chromo-


some with a translocation is called a derivative chromosome. For
the previous example, chromosomes 12 and 21 are referred to
as der(12) and der(21). Deletions are written with the abbre-
viation del preceding the chromosome. A deletion of the long
arm of chromosome 5 at band 31 is written as del(5)(q31).
No spaces are entered in these designations except between Figure 31-16 Hypodiploid karyotype with 36 chromosomes (arrows indi-
abbreviations.17 cate a missing chromosome).

CHROMOSOME ABNORMALITIES
cell divisions, meiosis I and meiosis II. The end result is a cell
There are many types of chromosome abnormalities, such as with 23 chromosomes, which is the haploid number (n).
deletions, inversions, ring formations, trisomies, and poly- In polyploidy, the chromosome number is higher than 46
ploidy. All these defects can be grouped into two major cate but is always an exact multiple of the haploid chromosome
gories: defects involving an abnormality in the number of number of 23. A karyotype with 69 chromosomes is called
chromosomes and defects involving structural changes in one triploidy (Figure 31-15). A karyotype with 92 chromosomes is
or more chromosomes. called tetraploidy.
In cancer, numerical abnormalities in the karyotype may be
Numeric Abnormalities classified further based on the modal number of chromosomes
Numeric abnormalities often are subclassified as aneuploidy in a neoplastic clone. Hypodiploid refers to a cell with fewer than
or polyploidy. Aneuploidy refers to any abnormal number of 46 chromosomes; near-haploid cells have from 23 up to approx-
chromosomes that is not a multiple of the haploid number (23 imately 34 chromosomes (Figure 31-16); hyperdiploid cells have
chromosomes). The common forms of aneuploidy in humans more than 46 chromosomes. High hyperdiploidy refers to a chro-
are trisomy (the presence of an extra chromosome) and mono- mosome number of more than 50.18 Finally, the term pseudo-
somy (the absence of a single chromosome). Aneuploidy is the diploid is used to describe a cell with 46 chromosome and
result of nondisjunction, the failure of chromosomes to sepa- structural abnormalities.
rate normally during cell division. Nondisjunction can occur
during either of the two types of cell division: mitosis or Structural Aberrations
meiosis. During normal mitosis, a cell divides once to produce Structural rearrangements result from breakage of a chromo-
two cells that are identical to the parent cell. In mitosis, each some region with loss or subsequent rejoining in an abnormal
daughter cell contains 46 chromosomes. Meiosis is a special combination. Structural rearrangements are defined as bal-
type of cell division that generates male and female gametes anced (no loss or gain of genetic chromatin) or unbalanced
(sperm and ova). In contrast to mitosis, meiosis entails two (gain or loss of genetic material). Structural rearrangements of
CHAPTER 31 Cytogenetics 453

Figure 31-17 A, Pericentric inversion. B, Paracentric inversion.

single chromosomes include inversions, deletions, isochromo- Duplication means partial trisomy for part of a chromosome.
somes, ring formations, insertions, translocations, and dupli- This can result from an unbalanced insertion or unequal cross-
cations. Inversions (inv) involve one or two breaks in a single ing over in meiosis or mitosis.
chromosome, followed by a 180-degree switch of the segment Translocations occur when there is breakage in two chromo-
between the breaks with no loss or gain of material. If the somes, and each of the broken pieces reunites with another
chromosomal material involves the centromere, the inversion chromosome. If chromatin is neither lost nor gained, the
is called pericentric. If the material that is inverted does not exchange is called a balanced reciprocal translocation. A reciprocal
include the centromere, the inversion is called paracentric translocation is balanced if all chromatin material is present.
(Figure 31-17). The loss or gain of chromatin material results in monosomy
Interstitial deletions arise after two breaks in the same chro- or trisomy for a segment of the chromosome, which is desig-
mosome and loss of the segment between the breaks. Terminal nated an unbalanced rearrangement.
deletions (loss of chromosomal material from the end of a Another type of translocation involving breakage and
chromosome) and interstitial deletions involve the loss of reunion near the centromeric regions of two acrocentric chro-
genetic material. The clinical consequence to the individual mosomes is known as a Robertsonian translocation. Effectively
with a deletion depends on the extent and location of the dele- this is a fusion between two whole chromosomes rather than
tion (Figure 31-18). exchange of material, as in a reciprocal translocation. These
Isochromosomes arise from either abnormal division of the translocations are among the most common balanced struc-
centromeres in which division is perpendicular to the long axis tural rearrangements seen in the general population, with a
of the chromosome rather than parallel to it or from breakage frequency of 0.09% to 0.1%.19 All five human acrocentric auto-
and reunion in chromatin adjacent to the centromere. Each somes (13, 14, 15, 21, and 22) are capable of forming a
resulting daughter cell has a chromosome in which the short Robertsonian translocation. In this case, the resulting balanced
arm or the long arm is duplicated. karyotype has only 45 chromosomes, which include the trans-
Ring chromosomes can result from breakage and reunion of located chromosomes (Figure 31-19).
a single chromosome with loss of chromosomal material
outside the break points. Alternatively, one or both telomeres
CANCER CYTOGENETICS
(chromosome ends) may join to form a ring chromosome
without significant loss of chromosomal material. Cancer cytogenetics is a field that has been built upon discovery
Insertions involve movement of a segment of a chromosome of nonrandom chromosome abnormalities in many types of
from one location of the chromosome to another location of cancer. In hematologic neoplasias, specific structural rearrange-
the same chromosome or to another chromosome. The ments are associated with distinct subtypes of leukemia that
segment is released as a result of two breaks, and the insertion have characteristic morphologic and clinical features. Cytoge-
occurs at the site of another break. netic analysis of malignant cells can help determine the
454 PART V Leukocyte Disorders

Figure 31-18 A, Interstitial deletion. B, Isochromosome. C, Ring chromosome. D, Insertion.

diagnosis and often the prognosis of a hematologic malig-


nancy, assist the oncologist in the selection of appropriate
therapy, and aid in monitoring the effects of therapy. Bone
marrow is the tissue most frequently used to study the cytoge-
1 2 3 4 5
netics of a hematologic malignancy. Unstimulated peripheral
blood and bone marrow trephine biopsy samples also may be
analyzed. Cytogenetic analysis of cancers involving other organ
6 7 8 9 10 11 12 systems can be performed using solid tissue obtained during
surgery or by needle biopsy. Chromosomal defects in cancer
include a wide range of numeric abnormalities and structural
13 14 15 16 17 18 rearrangements as discussed earlier (Table 31-1).
Cancer results from multiple and sequential genetic muta-
tions occurring in a somatic cell. At some juncture, a critical
19 20 21 22 X Y mutation occurs, and the cell becomes self-perpetuating or
Figure 31-19 Balanced Robertsonian translocation between chromo- clonal. A clone is a cell population derived from a single pro-
somes 14 and 21, 45,XY,der(14;21)(q10;q10). (Courtesy the Cytogenetics genitor.17 In cytogenetics, a clone exists if two or more cells
Laboratory, Indiana University School of Medicine, Indianapolis, Ind.) contain the same structural abnormality or supernumerary
CHAPTER 31 Cytogenetics 455

TABLE 31-1 Common Translocations in Hematopoietic


BOX 31-1 Acute Myeloid Leukemia (AML) with
and Lymphoid Neoplasia and Sarcoma
Recurrent Genetic Abnormalities
Tumor Type Karyotype Genes AML with t(8;21)(q22;q22);RUNX1/RUNX1T1
Myeloid Leukemias AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22);CBFB/MYH11
CML (and pre-B-ALL) t(9;22)(q34;q11.2) BCR/ABL1 Acute promyelocytic leukemia with t(15;17)(q22;q12);PML/RARA
Also see Box 31-1 AML with t(9;11)(p22;q23);MLLT3/MLL
AML with t(6;9)(p23;q34);DEK/NUP214
AML with inv(3)(q21q26.2) or t(3;3)(q21;q26.2);RPN1/EVI1
B Cell Leukemias /Lymphomas
AML (megakaryoblastic) with t(1;22)(p13;q13);RBM15/MKL1
B lymphoblastic leukemia t(12;21)(p13;q22) ETV6/RUNX1
AML with normal chromosomes and mutated NPM1
t(1;19)(q23.3;p13.3) PBX1/TCF3
AML with normal chromosomes and mutated CEBPA
t(4;11)(q21;q23) AF4/MLL
Burkitt lymphoma t(8;14)(q24;q32.3) MYC/IGH Modified from Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classifica-
t(2;8)(p12;q24) IGK/MYC tion of tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France,
t(8;22)(q24;q11.2) MYC/IGL 2008, IARC Press.
Mantle cell lymphoma t(11;14)(q13;q32.3) CCND1/IGH
Follicular lymphoma t(14;18)(q32.3;q21.3) IGH/BCL2
Diffuse large B cell t(3;14)(q27;q32.3) BCL6/IGH Leukemia
lymphoma Leukemias are clonal proliferations of malignant leukocytes
Lymphoplasmacytic t(9;14)(p13.2;q32.3) PAX5/IGH that arise initially in the bone marrow before disseminating to
lymphoma the peripheral blood, lymph nodes, and other organs. They
MALT lymphoma t(14;18)(q32.3;q21) IGH/MALT1 are broadly classified by the type of blood cell giving rise to
t(11;18)(q22;q21) BIRC3/MALT1 the clonal proliferation (lymphoid or myeloid) and by the
t(1;14)(p22;q32.3) BCL10/IGH clinical course of the disease (acute or chronic). The four main
leukemia categories are acute lymphoblastic leukemia (ALL),
T Cell Leukemias/Lymphomas acute myeloid leukemia (AML), chronic lymphocytic leukemia
T lymphoblastic leukemia del(1)(p32p32) STIL/TAL1 (CLL), and chronic myelogenous leukemia (CML). The World
t(7;11)(q35;p13) TRB/LMO2 Health Organization (WHO) classification for myeloid malig-
ALCL t(2;5)(p23;q35) ALK/NPM1 nancies has categorized AML into seven subtypes: AML with
recurrent genetic abnormalities; AML with myelodysplasia-
Sarcomas and Tumors of Bone and Soft Tissue related changes; therapy-related myeloid neoplasms; AML not
Alveolar t(2;13)(q36.1;q14.1) PAX3/FOXO1A otherwise specified; myeloid sarcoma; myeloid proliferations
rhabdomyosarcoma t(1;13)(p36.2;q14.1) PAX7/FOXO1A related to Down syndrome; and blastic plasmacytoid dendritic
Ewing sarcoma/PNET t(11;22)(q24;q12) FLI1/EWSR1 cell neoplasm (see Chapter 36).20 AML with recurrent genetic
t(21;22)(q22.3;q12) ERG/EWSR1 abnormalities is a classification based on the cytogenetic
t(7;22)(p22;q12) ETV1/EWSR1 abnormalities observed (Box 31-1). Some of the divisions of
Clear cell sarcoma t(12;22)(q13;q12) ATF1/EWSR1 the French-American-British (FAB) classification21 are included
Myxoid liposarcoma t(12;16)(q13;p11.2) DDIT3/FUS in the not otherwise classified category. The WHO has clas-
t(12;22)(q13;q12) DDIT3/EWSR1 sified lymphoid leukemias by precursor cell type, B or T (see
Synovial sarcoma t(X;18)(p11.2;q11.2) SSX1 or SSX2/SYT Chapter 36).
Alveolar soft part sarcoma t(X;17)(p11.2;q25) TFE3/ASPSCR1
Chronic Myelogenous Leukemia
ALCL, Anaplastic large cell leukemia; ALL, acute lymphoblastic leukemia; AML, acute The first malignancy to be associated with a specific chromo-
myeloid leukemia; CML, chronic myelogenous leukemia; CMML, chronic myelomono-
cytic leukemia; MALT, mucosa-associated lymphoid tissue; PNET, primitive neuro some defect was CML, in which approximately 95% of patients
ectodermal tumor. were found to have the Philadelphia chromosome, t(9;22)
(q34;q11.2) by G-banded analysis.22,23 The Philadelphia chro-
marker chromosome or if three or more cells are missing the mosome (derivative chromosome 22) is characterized by a
same chromosome. The primary aberration or stemline of a balanced translocation between the long arms of chromo-
clone is a cytogenetic abnormality that is frequently observed somes 9 and 22. At the molecular level, the gene for ABL1, an
as the sole abnormality associated with the cancer. The second- oncogene on chromosome 9, joins a gene on chromosome 22
ary aberration or sideline includes abnormalities additional to named BCR. The result of the fusion of these two genes is a
the primary aberration.17 In chronic myelogenous leukemia, new fusion protein of about 210 kD with growth-promoting
the primary aberration is the Philadelphia chromosome result- capabilities that override normal cell regulatory mechanisms
ing from a translocation between chromosomes 9 and 22, (Figures 31-20 and 31-21) (see Chapter 34).24 The fusion
t(9;22)(q34;q11.2). A sideline of this clone would include protein activates tyrosine kinase signaling to drive proliferation
secondary abnormalities, such as trisomy for chromosome 8, of the cell. This signaling can be blocked by imatinib mesylate
written as +8,t(9;22)(q34;q11.2). (Gleevec; Novartis Pharmaceuticals, East Hanover, N.J.).25
456 PART V Leukocyte Disorders

BCR/ABL1

MLL

Figure 31-20 Normal bone marrow interphase cell hybridized with the
BCR (green) and ABL (red) genes (Abbott Molecular, Des Plaines, Ill.). The Figure 31-22 Bone marrow metaphase cell with fusion MLL signal on
two red and two green signals represent the genes on the normal chromo- the normal chromosome 11 and split red and green signals on the translo-
somes 9 and 22. (Courtesy the Cytogenetics Laboratory, Indiana University cated chromosomes, representing a disruption of the MLL gene. (Courtesy
School of Medicine, Indianapolis, Ind.) the Cytogenetics Laboratory, Indiana University School of Medicine, India-
napolis, Ind.)
BCR/ABL1
MLL

Figure 31-23 Bone marrow interphase cell with a fusion signal and
split red and green signals from the MLL gene. (Courtesy the Cytogenetics
Laboratory, Indiana University School of Medicine, Indianapolis, Ind.)
Figure 31-21 Abnormal bone marrow interphase cell with one BCR
(green) and one ABL (red) signal (Abbott Molecular, Des Plaines, Ill.) repre- blood specimen to search for chromosomally abnormal cells.
senting the untranslocated chromosomes and two fusion signals from the This is an important advantage of FISH technology.
derivative chromosomes 9 and 22. (Courtesy the Cytogenetics Laboratory,
Indiana University School of Medicine, Indianapolis, Ind.) Acute Leukemia
The Philadelphia chromosome is also observed in acute leuke-
Patient response to imatinib can be monitored by cytogenetic mia. It is seen in about 20% of adults with ALL, 2% to 5% of
analysis and FISH. At diagnosis, the characteristic karyotype is children with ALL, and 1% of patients with AML.26-28 In child-
the presence of the Philadelphia chromosome in all cells ana- hood ALL, chromosome number is critical for predicting the
lyzed. After treatment for several months with imatinib, the severity of the leukemia. Children whose leukemic cells contain
karyotype typically has a mixture of abnormal and normal cells more than 50 chromosomes have the best prognosis for com-
indicating patient response to therapy. Complete response is plete recovery with therapy. Recurring translocations observed
defined as a bone marrow karyotype with only normal cells. in ALL include t(4;11)(q21;q23), t(12;21)(p13;q22), and
Often, therapeutic response is monitored using peripheral t(1;19)(q23;p13.3). Each translocation is associated with a
blood instead of a bone marrow aspirate. In contrast to the prognostic outcome and assists oncologists in determining
bone marrow, the peripheral blood does not contain spontane- patient therapy. The t(4;11) translocation is the one most com-
ously dividing cells. As a result, chromosomal analysis of a monly found in infants with acute lymphoblastic leukemia.
specimen of unstimulated peripheral blood often is unsuccess- Rearrangements of the AF4 gene on chromosome 4 and the
ful because of the absence of dividing cells. In these cases, FISH MLL gene on chromosome 11 occur in this translocation.29,30
with probes for the specific abnormality may be performed on Disruption of the MLL gene is seen in both ALL and AML.
200 or more interphase (nondividing) cells of the peripheral (Figures 31-22 and 31-23).
CHAPTER 31 Cytogenetics 457

HER2

Figure 31-24 Bone marrow metaphase chromosome 15 and 17 homo-


logues showing a translocation between the long arms of chromosomes 15
and 17, t(15;17)(q22;q21), diagnostic of acute promyelocytic leukemia. The
abnormal chromosomes are on the right. (Courtesy the Cytogenetics Labora-
tory, Indiana University School of Medicine, Indianapolis, Ind.)
Figure 31-26 Normal interphase nuclei from a paraffin-embedded tissue
section hybridized with probes for HER2 (red) and the chromosome 17
centromere (green) (Abbott Molecular, Des Plaines, Ill.). Two green and two
red signals are seen per cell.

HER2

Figure 31-25 Bone marrow karyogram for a patient with acute myeloid
leukemia showing a translocation, t(9;22)(q34;q11.2), and an inverted chro-
mosome 16, inv(16)(p13.1q22). (Courtesy the Cytogenetics Laboratory,
Indiana University School of Medicine, Indianapolis, Ind.)

The AMLs are subdivided into several morphologic


classifications ranging from M0 to M7 according to the FAB Figure 31-27 Tissue section from a breast cancer demonstrating ampli-
classification (see Chapter 36).31,32 Characteristic chromosome fication of HER2. The tissue was hybridized with fluorescence in situ hybrid-
translocations are associated with some subgroups and were ization probes for HER2 (red) and the chromosome 17 centromere probe
incorporated into the WHO classification. Among them is a (green) (Abbott Molecular, Des Plaines, Ill.). The number of HER2 signals
exceeds the number of centromere signals, which indicates selective ampli-
translocation between the long arm of chromosome 8 and the
fication of HER2.
long arm of chromosome 21, t(8;21)(q22;q22), which is rep-
resentative of AML with maturation. Acute promyelocytic leu-
kemia is associated with a translocation between the long arms Solid Tumors
of chromosomes 15 and 17, t(15;17)(q24;q21) (Figure 31-24). Just as recurring structural and numeric chromosome defects
A pericentric inversion of chromosome 16, inv(16)(p13.2q22), have been observed in the hematologic malignancies, a wide
is seen in AML with increased eosinophils. The inversion jux- range of abnormalities have also been found in solid tumors.
taposes core binding factor beta (CBFB) gene with the myosin Most of these abnormalities confer a proliferative advantage
heavy chain gene (MYH11) to form a new fusion protein on the malignant cell and serve as useful prognostic indicators.
(Figure 31-25).33 These recurring translocations have enabled Amplification of the gene HER2 (also called ERBB2) on chro-
researchers to localize genes important for cell growth and mosome 17, a transmembrane growth factor receptor, is associ-
regulation. As with acute lymphoblastic leukemia, the particu- ated with an aggressive form of invasive breast cancer.34,35 FISH
lar translocation often predicts patient prognosis and response with probes for the HER2 gene and an internal control (17
to therapy. Understanding the molecular consequences of the centromere) can determine if there is gene amplification in the
cytogenetic mutations, such as the BCR/ABL translocation, pro- tumor (Figure 31-26).36 If FISH testing shows amplification to
vides the fundamental information for the development of be present, the patient is eligible for targeted therapy with a
targeted therapies. monoclonal antibody, trastuzumab (Figure 31-27).37 FISH for
458 PART V Leukocyte Disorders

HER2 typically is performed on tissue sections from the


paraffin-embedded tumor block.
Array Comparative Genomic
Hybridization (aCGH)

ARRAY COMPARATIVE
Patients DNA Reference DNA
GENOMIC HYBRIDIZATION
Array comparative genomic hybridization (aCGH) is a new
 fluorescence-based molecular technique for analyzing genomic
DNA for unbalanced genetic alterations. In this method patient
DNA and a normal reference DNA are differently labeled, one
with a green fluorophore and one with a red fluorophore.
BAC/Oligo DNAs The two DNAs are mixed together and hybridized to an array
platform that contains spotted DNA representing the target(s)
of interest. Regions of imbalance (copy gain or copy loss)
in the patient specimen are assessed relative to the fluores-
cence signal of the reference DNA when hybridized to the
normal DNA of the array (Figure 31-28).38,39 This technique is
presently used primarily for diagnosis of constitutional (inher-
ited) disorders but emerging applications for cancer are in
development.

Copy loss Copy gain

Figure 31-28 Diagram of the DNA labeling and hybridization for array
comparative genomic hybridization.

SUMMARY
Cytogenetics is the study of chromosome structure and Tissues used for chromosome analysis include bone marrow cells
inheritance. and peripheral blood lymphocytes, amniotic fluid, nonneoplastic
Chromosome disorders may be structural or numeric, involving tissue, and tumors.
the rearrangement or the loss or gain of a piece of a chromosome A normal cell contains 46 chromosomes, which includes 2 sex
or the entire chromosome. chromosomes.
Nonrandom chromosome abnormalities are associated with Defects in chromosomes can be categorized as numeric or struc
cancer. tural. Numeric abnormalities can be subclassified as aneuploidy
A chromosome is composed of a complementary double helix of and polyploidy.
DNA. Attached to the backbone of deoxyribose are adenine (A), Structural rearrangements include inversions, deletions, isochro
guanine (G), cytosine (C), and thymine (T). mosomes, ring formations, insertions, translocations, and
During mitosis, cells can be chemically treated to arrest cell pro duplications.
gression in metaphase so that chromosomes can be analyzed. Specific structural rearrangements are associated with distinct
Q banding differentiates chromosomes into bands of different subtypes of leukemias, and analysis may assist in diagnosis,
widths and relative brightnesses, revealing a banding pattern prognosis, and monitoring of therapy. Solid tumors also may be
unique to each individual chromosome. analyzed using cytogenetics.
Other stains used to identify chromosomes may require pre
treatment of the slide for analysis. These include G banding, C Now that you have completed this chapter, go back and
banding, and AG-NOR banding. read again the case study at the beginning and respond
FISH, a molecular cytogenetic technique, uses DNA or RNA probes to the questions presented.
and fluorescence microscopy to identify individual chromosomes.
Metaphase and interphase cells can be analyzed by FISH.
CHAPTER 31 Cytogenetics 459

R E V I E W Q UESTIONS
1. G-banding refers to the technique of staining chromosomes: 6. Which of the following describes a chromosomal
a. To isolate those in the G group (i.e., chromosomes 21 deletion?
and 22) a. Point mutation resulting in a single amino acid
b. In the G0 or resting stage substitution
c. Using Giemsa stain b. Transfer of genetic material from one chromosome to
d. To emphasize areas high in guanine residues another
c. Loss of genetic material from a chromosome that
2. Which of the following compounds is used to halt mitosis does not appear on any other chromosome
in metaphase for chromosome analyses? d. Duplication of a chromosome resulting in 3n of that
a. Imatinib genetic material
b. Fluorescein
c. Trypsin The chromosome analysis performed on a patients leuke-
d. Colchicine mic cells is reported as 47, XY,+4,del(5)(q31)[20]. Answer
questions 7 to 9 based on this description.
3. One arm of a chromosome has 30 bands. Which band
would be nearest the centromere? 7. This patients cells have which of the following mutations?
a. Band 1 a. Loss of the entire number 31 chromosome
b. Band 15 b. Loss of the entire number 5 chromosome
c. Band 30 c. Loss of a portion of the short arm of chromosome 4
d. Loss of a portion of the long arm of chromosome 5
4. Which of the following is not an advantage of the use of
FISH? 8. What other mutation is present in this patients cells?
a. It can be used on nondividing cells. a. Polyploidy
b. It can be used on paraffin-embedded tissue. b. Tetraploidy
c. It can detect mutations that do not result in abnormal c. An extra chromosome 4
banding patterns. d. Four copies of chromosome 5
d. It must be performed on dividing cells.
9. This patients leukemic cells demonstrate:
5. Which of the following types of mutations would likely a. Structural chromosomal defects only
not be detectable with cytogenetic banding techniques? b. Numeric chromosomal defects only
a. Point mutation resulting in a single amino acid c. Both structural and numeric chromosomal defects
substitution
b. Transfer of genetic material from one chromosome to 10. Aneuploidy describes the total chromosome number:
another a. That is a multiple of the haploid number
c. Loss of genetic material from a chromosome that b. That reflects a loss or gain of a single chromosome
does not appear on any other chromosome c. That is diploid but has a balanced deletion and
d. Duplication of a chromosome resulting in 3n of that duplication of whole chromosomes
genetic material d. In gametes; diploid is the number in somatic cells

REFERENCES
1. International Human Genome Sequencing Consortium: 6. Earnshaw WC: Mitotic chromosome structure. Bioessays
Finishing the euchromatic sequence of the human genome. 9:147-150, 1988.
Nature 431:931-945, 2004. 7. Olins AL, Olins DE: Spheroid chromatin units. Science
2. Benn P: Prenatal diagnosis of chromosomal abnormalities 183:330-332, 1974.
through amniocentesis. In Milunsky A, Milunsky JM, editors: 8. Oudet P, Gross-Bellard M, Chambon P: Electron microscopic
Genetic disorders and the fetus, ed 6, Baltimore, 2010, Wiley- and biochemical evidence that chromatin structure is a
Blackwell, pp 194-195. repeating unit. Cell 4:281-300, 1975.
3. Bou J, Bou A, Lazar P: Retrospective and prospective epide- 9. Rattner JB: Chromatin hierarchies and metaphase chro
miological studies of 1500 karyotyped spontaneous human matin structure. In Adolph KW, editor: Chromosomes and
abortions. Teratology 12:11-26, 1975. chromatin, vol 3, Boca Raton, Fla., 1988, CRC Press,
4. Rowley JD: Chromosomal translocations: revisited yet again. p 30.
Blood 112:2183-2189, 2008. 10. Tijo JH, Levan A: The chromosome number of man. Hereditas
5. Watson JD, Crick FHC: Molecular structure of nucleic acids: 42:1-6, 1956.
a structure for deoxyribose nucleic acid. Nature 171:737-738, 11. Hsu TC: Human and mammalian cytogenetics. New York, 1979,
1953. Springer-Verlag, pp 6-7.
460 PART V Leukocyte Disorders

12. Nowell PC: Phytohemagglutinin: an initiator of mitosis in childhood acute lymphoblastic leukemia. Blood 70:948-953,
culture of normal human leukocytes. Cancer Res 20:462-466, 1987.
1960. 28. Crist W, Carroll A, Shuster J, et al: Philadelphia chromosome
13. Farnes P, Barker BE, Brownhill LE, et al: Mitogenic activity positive acute lymphoblastic leukemia: clinical and cytoge-
of Phytolacca americana (pokeweed). Lancet 2:1100-1101, netic characteristics and treatment outcome. A Pediatric
1964. Oncology Group study. Blood 76:489-494, 1990.
14. Caspersson T, Zech L, Modest EJ, et al: Chemical differentia- 29. Morrissey J, Tkachuk DC, Milatovich A, et al: A serine/
tion with fluorescent alkylating agents in Vicia faba meta- proline-rich protein is fused to HRX in t(4;11) acute leuke-
phase chromosomes. Exp Cell Res 58:128-140, 1969. mias. Blood 81:1124-1131, 1993.
15. Yunis JJ: Cytogenetics. In Henry JB, editor: Clinical diagnosis 30. Hayne CC, Winer E, Williams T, et al: Acute lymphoblastic
and management by laboratory methods, ed 16. Philadelphia, leukemia with a 4;11 translocation analyzed by multi-modal
1979, Saunders, p 825. strategy of conventional cytogenetics, FISH, morphology,
16. Plattner R, Heerema NA, Yurov YB, et al: Efficient identifica- flow cytometry, molecular genetics and review of the litera-
tion of marker chromosomes in 27 patients by stepwise ture. Exp Mol Pathol 81:62-71, 2006.
hybridization with alpha satellite DNA probes. Hum Genet 31. Bennett JM, Catovsky D, Daniel MT, et al: Proposed revised
91:131-140, 1993. criteria for the classification of acute myeloid leukemia: a
17. Shaffer LG, Slovak ML, Campbell LJ, editors: ISCN (2009): report of the French-American-British Cooperative Group.
an international system for human cytogenetic nomenclature, Ann Intern Med 103:620-625, 1985.
Basel, Switzerland, 2009, S Karger. 32. Bennett JM, Catovsky D, Daniel MT, et al: Criteria for the
18. Moorman AV, Richards SM, Martineau M, et al: Outcome diagnosis of acute leukemia of megakaryocytic lineage (M7):
heterogeneity in childhood high-hyperdiploid acute lympho- a report of the French-American-British Cooperative Group.
blastic leukemia. Blood 102:2756-2762, 2003. Ann Intern Med 103:460-462, 1985.
19. Turleau C, Chavin-Colin F, de Grouchy J: Cytogenetic inves- 33. Claxton DF, Liu P, Hsu HB, et al: Detection of fusion tran-
tigation in 413 couples with spontaneous abortions. Eur J scripts generated by the inversion 16 chromosome in acute
Obstet Gynecol Reprod Biol 9:65-74, 1979. myelogenous leukemia. Blood 83:1750-1756, 1994.
20. Swerdlow SH, Campo E, Harris NL, et al, editors: WHO 34. Slamon DJ, Godolphin W, Jones LA, et al: Studies of the
classification of tumours of haematopoietic and lymphoid tissues, HER-2 proto-oncogene in human breast and ovarian cancer.
ed 4, Lyon, France, 2008, IARC Press. Science 244:707-712, 1989.
21. Bennett JM, Catovsky D, Daniel MT, et al: Proposals for clas- 35. Perez EA, Roche PC, Jenkins RB, et al: HER2 testing in patients
sification of the acute leukaemias. French-American-British with breast cancer: poor correlation between weak positivity
(FAB) Co-operative Group. Br J Haematol 33:451-458, 1976. by immunohistochemistry and gene amplification by fluores-
22. Nowell PC, Hungerford DA: A minute chromosome in cence in situ hybridization. Mayo Clin Proc 77:148-154, 2002.
human chronic granulocytic leukemia. Science 132:1497 36. Wolff AC, Hammond E, Schwartz JN, et al: American Society
(abstract), 1960. of Clinical Oncology/College of American Pathologists
23. Rowley JD: A new consistent chromosomal abnormality in guideline recommendations for human epidermal growth
chronic myelogenous leukemia identified by quinacrine fluo- factor receptor 2 testing in breast cancer. J Clin Oncol
rescence and Giemsa staining. Nature 243:290-293, 1973. 25(1):118-145, 2007.
24. Ben-Neriah Y, Daley GQ, Mes-Masson A, et al: The chronic 37. Slamon DJ, Leyland-Jones B, Shak S, et al: Use of chemo-
myelogenous leukemiaspecific P210 protein is the product therapy plus a monoclonal antibody against HER2 for meta-
of the BCR/ABL hybrid gene. Science 233:212-214, 1986. static breast cancer that overexpresses HER2. N Engl J Med
25. Druker BJ, Tamura S, Buchdunger E, et al: Effects of a selective 344:783-792, 2001.
inhibitor of the Abl tyrosine kinase on the growth of BCR-ABL 38. Stankiewicz P, Beaudet AL: Use of array CGH in the
positive cells. Nat Med 2:561-566, 1996. evaluation of dysmorphology, malformations, developmen-
26. Third International Workshop on Chromosomes in Leuke- tal delay, and idiopathic mental retardation. Curr Opin Genet
mia: Chromosomal abnormalities in acute lymphoblastic Dev 17:182-192, 2007.
leukemia: structural and numerical changes in 234 cases. 39. Baldwin EL, Lee JY, Blake DM, et al: Enhanced detection
Cancer Genet Cytogenet 4:101-110, 1981. of clinically relevant genomic imbalances using a targeted
27. Ribeiro RC, Abromowitch M, Raimondi SC, et al: Clinical plus whole genome oligonucleotide microarray. Genet Med
and biologic hallmarks of the Philadelphia chromosome in 10:415-429, 2008.
Molecular Diagnostics in
the Clinical Laboratory
32
Mark E. Lasbury

OUTLINE OBJECTIVES
Structure and Function After completion of this chapter, the reader will be able to:
of DNA
The Central Dogma: DNA 1. Describe the structure of DNA, including the com 7. Discuss DNA isolation.
to RNA to Protein position of a nucleotide, the double helix, strand 8. Explain the purpose of polymerase chain reaction
DNA at the Molecular orientation, and the antiparallel complementary (PCR), reverse transcription PCR, and nucleic acid
Level characteristic. hybridization.
Transcription and 2. Predict the nucleotide sequence of a complementary 9. Explain DNA sequencing.
Translation strand of DNA or RNA given the nucleotide sequence 10. Compare and contrast the methods for detecting
DNA Replication and of a DNA template. amplified target DNA: (1) gel electrophoresis using
the Cell Cycle 3. Explain the relationship between DNA structure and either ethidium bromide or autoradiography, (2)
Molecular Diagnostic protein production. restriction fragment length polymorphism, and (3)
Testing Overview
4. Explain the relationship between the cell cycle and probe hybridization techniques such as Southern
Nucleic Acid Isolation
tumor progression. blotting.
(Extraction)
Isolating DNA from 5. Discuss the process of DNA replication, including 11. Interpret an agarose gel electrophoresis result for the
Clinical Specimens replication origin, replication fork, primase, primer, factor V Leiden mutation test and a nucleic acid
Isolating RNA from DNA polymerase, Okazaki fragments, leading strand, hybridization result for a B- and T-cell gene rear
Clinical Specimens and lagging strand. rangement test.
Amplification of Nucleic 6. Determine the appropriate patient specimen required 12. Describe the principle of real-time quantitative PCR.
Acids by Polymerase for DNA isolation to identify an inherited or somatic 13. Discuss the use of real-time PCR for monitoring
Chain Reaction mutation. minimal residual disease.
Polymerase Chain
Reaction for Amplifying
DNA
CASE STUDY*
Reverse Transcription
Polymerase Chain After studying this chapter, the reader should be able to respond to the following case study:
Reaction for Amplifying
A 42-year-old man came to the emergency room complaining of pain behind his right knee. He
RNA
Detection of Amplified had observed swelling below the knee for the previous 2 days. The patient was in no apparent
DNA distress and was experiencing no chest pain, shortness of breath, dyspnea, or hemoptysis. The
Gel Electrophoresis patient reported no history of trauma except for a right femur break 20 years before. He reported
Restriction Endonuclease that he did not smoke, used alcohol occasionally, was taking no medications, and was in good
Methods general health. He was not overweight and had not experienced any malaise, weight loss, arthralgia,
Nucleic Acid Hybridization or melena. He jogs 5 miles daily. Five years previously, he experienced an episode of deep vein
and the Southern thrombosis (DVT) in his right lower leg and was treated with intravenous heparin followed by oral
Blotting warfarin (Coumadin) for 3 months. Subsequent to treatment, he experienced occasional pain
Hybridization Labels behind both knees, which he treated with aspirin. He noted that his mother had been diagnosed
DNA Sequencing
with carpal tunnel syndrome and had developed DVT, for which she has been taking oral warfarin
Real-Time Polymerase
Continued
Chain Reaction
Minimal Residual Disease
in Leukemia *This case was provided by George A. Fritsma, MS, MLS, manager, The Fritsma Factor: Your Interactive
Infectious Disease Load Hemostasis Resource, http://www.fritsmafactor.com, sponsored by Precision BioLogic, Inc., Cambridge, Nova
Current Developments Scotia.

461
462 PART V Leukocyte Disorders

CASE STUDYcontd
After studying this chapter, the reader should be able to respond to the following case study:

for 15 years. There is no family history of collagen disease. the factor V Leiden mutation test initially done by the mole
The patients job requires frequent long airplane flights. He cular scientist. The scientists supervisor reviewed the gel
flies first class and walks around occasionally during the long electrophoresis results and requested that the scientist repeat
flights. His leg pain began 1 week after a flight to Europe. the analysis. Figure 32-2 represents the repeated analysis.
On physical examination, the patient had no evidence of 1. What type of specimen is appropriate when analyzing
rash or oral ulcers. No petechiae or purpura were noted. He DNA for a hereditary mutation?
had mild pretibial pitting edema. His right leg measured 2. Examine the first gel electrophoresis result (see Figure
36.5cm at 25cm distal to the superior aspect of the patella, 32-1). Are the correct controls present?
whereas his left leg measured 33.5cm in the same location. 3. Examine the second gel electrophoresis result (see Figure
CBC findings were unremarkable, and both the prothrombin 32-2). What band sizes appear in the patients sample?
time and activated partial prothrombin time were within the 4. What band sizes are expected for an individual who is
reference ranges. Doppler ultrasonography revealed complete homozygous for the factor V Leiden mutation, heterozy-
occlusion of the distal superficial femoral vein, anterior tibial gous for the mutation, and free of the mutation?
vein, and popliteal vein. The diagnosis was DVT without 5. Why did the laboratory request that the blood sample to
pulmonary emboli. The patient was hospitalized, and a be redrawn?
heparin drip was started. The hematologist ordered a factor 6. Why was a repeat analysis requested?
V Leiden analysis. The initial specimen received was drawn 7. Does this patient have factor V Leiden?
into a red-topped tube. The laboratory scientist requested a
redraw using a lavender-topped tube. The patient was still
hospitalized, which allowed the collection of a blood speci-
men in an EDTA tube. Figure 32-1 illustrates the results of

Figure 32-1 Results of the initial factor V Leiden mutation test done by Figure 32-2 Results of the repeated factor V Leiden mutation test
the molecular scientist described in the case study. Lane A, molecular size performed by the molecular scientist in the case study. Lane A, molecular
marker (ladder); B, positive control; C, patients sample; D, negative control. size marker; B, positive control; C, patients sample; D, negative control;
The expected banding pattern on an agarose gel for the factor V mutation E, no-DNA control.
test is as follows: B, homozygous for the factor V mutation: 141 and 82bp;
C, heterozygous for the factor V mutation: 141, 104, and 82bp; D, no
factor V mutation: 104 and 82bp. The band at 37bp is barely visible and
is difficult to detect on an agarose gel. However, this band is not essential
for interpreting the results.
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 463

M
olecular biology techniques enhance the diagnostic
teams ability to predict or identify an increasing BOX 32-2 Inherited Hematologic Disorders Detected
number of diseases in the clinical laboratory. by Molecular Diagnostic Methods
Molecular techniques also enable clinicians to monitor disease Hemoglobinopathies
progression during treatment and to make accurate prognoses. Sickle cell anemia
The short interval required to perform molecular diagnostic Hemoglobin C disease
tests and analyze their results is an additional positive aspect Hemoglobin SC disease
of this type of testing, resulting in more efficient patient man- Hemoglobin E disease
agement, especially in cases of infection. The three main areas Hemoglobin D disease
of hematopathologic molecular testing include detection of -Thalassemias
chromosomal translocations in hematologic malignancies -Thalassemias
(Box 32-1) and inherited hematologic disorders (Box 32-2), Hereditary persistence of fetal hemoglobin
identification of hematologically important infectious diseases Coagulopathies
(Box 32-3), and monitoring of minimal residual disease after Hemophilia A
cancer treatment. Hemophilia B
Thrombophilia
Coagulation factor V Leiden mutation
STRUCTURE AND FUNCTION OF DNA Prothrombin G20210A mutation
The Central Dogma: DNA to RNA to Protein MTHFR C677T
Much of the stored information needed to carry out cell pro- MTHFR A1298T
cesses resides in deoxyribonucleic acid (DNA); therefore, Warfarin sensitivity
proper cellular storage, maintenance, and replication of DNA CYP2C9*2
are necessary to ensure homeostasis. Since molecular testing CYP2C9*3
takes advantage of DNA structure and replication, a review of VKORC1
molecular biology is helpful. Erythrocytic disorders
The central dogma in genetics is that information stored in Lipid storage diseases
the DNA is replicated to daughter DNA, transcribed to messenger Neutrophil disorders
ribonucleic acid (mRNA), and translated into a functional Modified from Crisan D: Pioneering advances in molecular hematology: use of
protein (Figure 32-3). This process is essential to carry out cel- nucleic-acid technology in hematologic disease diagnosis and monitoring
lular functions while preserving a record of the stored informa- molecular pathology, part 4, MLO Med Lab Obs 27(10):48-56, 1995.
tion. In eukaryotes, the initial DNA sequence is composed of
exons separated by untranslated introns. The introns are enzy-
matically excised during transcription from DNA to RNA, and
the mature mRNA sequence is then translated. Translation is
an enzymatic process wherein mRNA three-member base
sequences called codons drive the addition of individual amino
BOX 32-3 Hematologically Important Pathogens
Detected by Molecular Diagnostic Methods
Parasitic pathogens
BOX 32-1 Hematologic Malignancies Possessing p53 Plasmodium
Alterations Filaria
B-cell acute lymphoblastic leukemia Babesia
B-cell prolymphoblastic leukemia Leishmania
T-cell acute lymphoblastic leukemia Trypanosoma
Chronic lymphocytic leukemia (CLL) Fungal pathogens
Hairy cell leukemia Bacterial pathogens
Atypical CLL Viral pathogens
Myelodysplastic neoplasms Viral pathogens associated with hemolytic anemia
Acute myeloid leukemia Parvovirus B19
Chronic myelogenous leukemia Cytomegalovirus
Diffuse large B-cell non-Hodgkin lymphoma Epstein-Barr virus
Splenic lymphoma with villous lymphocytes Human immunodeficiency virus types 1 and 2
Non-Hodgkin lymphoma Human T-cell lymphoma virus type 1
Mantle cell lymphoma
Richter syndrome Modified from Paessler M, Bagg A: Use of molecular techniques in the analysis
of hematologic diseases. In Hoffman R, Benz EJ Jr, Shattil SJ, et al, editors:
Modified from Peller S, Rotter V: TP53 in hematological cancer: low incidence of Hematology: basic principles and practice, ed 4, Philadelphia, 2005, Churchill
mutations with significant clinical relevance, Hum Mutat 21:277-284, 2003. Livingstone, pp 2713-2726.
464 PART V Leukocyte Disorders

Nucleus

Cytoplasm

Figure 32-3 RNA polymerase transcribes DNA into a primary RNA transcript. The introns in the primary RNA transcript are excised and the exons are
spliced. The spliced messenger RNA (mRNA) enters the cytoplasm of the cell. The ribosomes translate the mRNA into protein.

acids to the growing peptide. The mature protein then carries DNA at the Molecular Level
out its cellular function, which may be structural or may DNA is a duplex molecule composed of two complementary
involve recognition, regulation, or enzymatic activity. hydrogen-bonded nucleotide strands (Figure 32-4). Deoxyribo-
The structural units that carry DNAs message are called nucleotides and ribonucleotides are the building blocks of
genes. The human -globin gene, part of the hemoglobin DNA and RNA, respectively. Each nucleotide is composed of a
molecule, provides a good example of replication and 5-carbon sugar (pentose), a nitrogenous base, and a phosphate
transcription, because it was one of the first sequenced and group (Figure 32-5). The numbers one prime (1) to five prime
demonstrates the result of aberrant sequence maintenance. A (5) designate the pentoses carbons. In DNA, the pentose is a
normal (or wild-type) -globin gene contains a sequence of ribose in which the hydroxyl group on the 2 carbon is replaced
bases that code for a -globin peptide of 146 amino acids. One by a hydrogen molecule, hence 2-deoxyribose. In RNA, the 2
inherited mutation changes a single DNA base. This is called a ribose retains the 2 hydroxyl group. The hydroxyl group
point mutation. The mutation occurs in the portion of the present on the 3 carbon of the sugar is crucial for polymeriza-
sequence that codes for the sixth amino acid of -globin. The tion of the nucleotide monomers to form the nucleic acid
mutation substitutes the amino acid valine for glutamine in strand.
the growing peptide. Valine modifies the overall charge, pro- The nitrogenous base is linked to the sugar by a glycosidic
ducing a protein that polymerizes in a low-oxygen environ- bond at the 1 carbon. Four different bases form DNA, but the
ment. This leads to sickled erythrocytes, circulatory ischemia, linkage to the sugar is the same for each. The phosphate group
and poor oxygen exchange between blood and tissues.1,2 A is linked to sugar at the 5 carbon by a phosphodiester bond.
mutation in one of the two copies (alleles) of this gene inher- The phosphate group is also crucial for addition of nucleotides
ited from the parents results in a heterozygous condition, or a to the growing polymer. A sugar, whether ribose or deoxyri-
sickle cell trait. In a heterozygote, the symptoms of the disease bose, linked to a nitrogenous base, but without a phosphate
are often unseen or are present only during times of physical group, is called a nucleoside. A nucleoside cannot be incorpo-
stress. If both alleles are mutated, there is overt homozygous rated into DNA, and neither can a nucleotide consisting of only
sickle cell disease, and the symptoms are severe. one phosphate group (deoxynucleotide monophosphate, or
Every active gene is translated. Human somatic cells contain dNMP). To be incorporated into a growing strand of DNA,
20,000 to 25,000 genes in 2meters of DNA.3,4 Significant the nucleotide must have three phosphate groups linked to
packing (see Chapter 31) takes place to reduce the volume of one another, referred to as the -, -, and -phosphates. The
the nucleic acid to the size of chromosomes. -phosphate is linked to the sugar.
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 465

Figure 32-4 DNA is a double-stranded helical macromolecule consisting of nucleotide subunits joined in sequence by deoxyribose molecules (pentagons)
and phosphate radicals (circles). The bases thymine (T), adenine (A), cytosine (C), and guanine (G) are illustrated in their standard pairs: thymine to adenine,
cytosine to guanine.

Creation of a phosphodiester bond between the 3 hydroxyl ladder, with the repeating sugar and phosphate groups forming
group of the existing strand and the 5 -phosphate of the the sides of the ladder and the bases forming the rungs. The
nucleotide monomer requires the protein enzyme DNA poly- pairing of a double-ringed purine on one strand with a single-
merase. This enzyme recognizes the hydroxyl group on the 3 ringed pyrimidine on the other maintains a consistent distance
carbon of the sugar and bonds the 3 hydroxyl group of one between the DNA strands. This makes DNA flexible, which
nucleotide with the -phosphate group of another (Figure allows the molecule to twist into a helix. Twisting stabilizes the
32-6). Polymerization of subsequent nucleotides forms a molecule and protects the bases from their environment.
DNA strand.
DNA consists of two strands that are antiparallel and comple- Transcription and Translation
mentary (Figure 32-7). One strand begins with a phosphate DNA provides a permanent set of instructions. The cellular
group attached to the 5 carbon of the first nucleotide oriented enzyme RNA polymerase transcribes the code. RNA polymerase
to the left and ends with the hydroxyl group on the 3 carbon recognizes starter sequences called promoters. Promoters lie
of the last nucleotide oriented to the right. This strand is in the upstream of coding sequences and bind RNA polymerase to
5-to-3 direction. The other strand runs in the 3-to-5 direc- separating DNA strands. The enzyme then slides along the
tion, or antiparallel. The nucleotide sequences composing DNA strand 3 to 5, reading the code and polymerizing
these strands provide the encoded messages of our genes. (assembling) the complementary ribonucleotides. As the com-
Therefore, the addition of nucleotides is highly regulated. plementary ribonucleotides form hydrogen bonds with the
One regulation mechanism arises from the complementary bases of the exposed DNA strand, the RNA polymerase creates
characteristic of the nucleotides. A nucleotides identity phosphodiester bonds to extend the single-stranded primary
depends on the type of nitrogenous base present on the tem- RNA transcript (Figure 32-10). If the nucleotide sequence of
plate. There are two categories of nitrogenous bases in nucleic the DNA strand is 3-CTAG-5, the primary RNA transcript is
acids, purines and pyrimidines (Figure 32-8). The bases adenine 5-GAUC-3.
(A) and guanine (G) are double-ringed purines, whereas thymine Primary mRNA segments are composed of introns and exons.
(T) and cytosine (C) are single-ringed pyrimidines. Adenine Introns are untranslated intervening sequences located within
forms hydrogen bonds at two points with thymine (A:T), the coding portions of genes. Their functions remain unclear,
whereas guanine forms hydrogen bonds at three points with although they may play a role in regulation of gene expres-
cytosine (G:C). If a strand has a 5-CTAG-3 sequence, the sion.5 Exons are the sequences that encode the gene product.
complementary nucleotides on the 3-to-5 strand are 3-GATC- Before mRNA can serve as a translation template, the introns
5. In RNA, the pyrimidine uracil (U) takes the place of thymine must be excised from the primary transcript and the exons
and forms hydrogen bonds with adenine. Hydrogen bonds adjoined. The mature mRNA is completed by the addition of
between A:T and G:C hold the strands together (Figure 32-9). a 5 cap and a tail of many repeated adenine nucleotides.6 The
RNA is most often single-stranded. mRNA leaves the nucleus and enters cytoplasmic ribosomes to
In addition to conferring identity to the nucleotide, the be translated.
nitrogenous bases assist in maintaining a constant width Ribosomes translate the mRNA code into a peptide
between the strands of a DNA molecule. DNA resembles a sequence. Complexes of proteins and structural ribosomal
466 PART V Leukocyte Disorders

Figure 32-5 A, The pentose sugar deoxyribose, a phosphate group, and a nitrogenous base compose a DNA nucleotide. The carbons of the deoxyribose
molecule are numbered 1 through 5. B, A nucleotide results from the formation of a phosphodiester bond between the nitrogenous base and the hydroxyl
group on the 1 carbon of deoxyribose and a glycosidic bond between the phosphate group and the hydroxyl group on the 5 carbon of deoxyribose.
C, A nucleotide illustrating the glycosidic and phosphodiester bonds.

RNAs (rRNAs) form both large and small ribosome subunits. step in translation is hydrogen bonding of the appropriately
Mature cytoplasmic mRNA is bound by the small ribosomal charged tRNA (with a bound methionine) to the start codon
subunit at the translation start site. At this point, another series of the mRNA. The appropriate tRNA is then bonded to the
of elements is introduced, transfer RNAs (tRNAs), each bound adjacent codon and a peptide bond is catalyzed between the
to its specific amino acid. Because there are 20 natural amino two amino acids. The peptide bond forms between the car-
acids, there are 20 tRNAs. Each tRNA has a specific nucleic acid boxyl terminus of the methionine in the existing peptide chain
sequence located at the point of interaction with the mRNA, and the amino terminus of the amino acid to be added. Hydro-
complementary to the nucleotide sequence of the mRNA. Each gen bonding of tRNAs to the codons and the formation of the
tRNA interacting sequence (anticodon) complements a specific peptide bonds are mediated by the ribosome. With addition of
three-nucleotide sequence (codon) of the mRNA. more amino acids, translation proceeds until a termination site
The mRNA codon AUG is the most common translation (nonsense codon) is reached. Three codons exist that do not
start site and codes for the amino acid methionine. The first code for any amino acid: UAA, UAG, and UGA. These
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 467

-phosphate -phosphate
O O

+ HO P O P OH
O O OH OH
HO P O P O
OH OH OH
-phosphate -phosphate -phosphate

Figure 32-6 The enzyme DNA polymerase catalyzes the reaction between the hydroxyl group on the 3 carbon of one nucleotide with the phosphate group
bound to the 5 carbon of the downstream nucleotide. The -phosphate group is split by the 3-OH, with release of the - and -phosphates.

Figure 32-8 The single-ringed pyrimidines thymine and cytosine and the
double-ringed purines adenine and guanine are the code-carrying nitroge
nous bases of DNA.

The cell cycle progresses through a sequence (Figure 32-11).


Interphase is made up of the G1, S, and G2 phases. During the
G1 phase, the cell grows rapidly and performs its cellular func-
tions. S phase is the synthesis stage, in which DNA is replicated.
The G2 phase is the period when the cell produces materials
essential for cell division. The M phase refers to mitosis, during
which two identical daughter cells are produced, each of which
Figure 32-7 DNA consists of two antiparallel and complementary strands. receives one entire set of the DNA that was replicated during S
One strand begins with a 5 phosphate group and ends with a 3 hydroxyl phase. Some cells exit the cell cycle during the G1 phase and
group. This strand is read in the 5-to-3 direction. The other strand begins enter a phase called G0. Cells in G0 normally do not reenter the
with a 3 hydroxyl group and ends with a 5 phosphate group. This strand is cell cycle and remain alive performing their function until
shown in the 3-to-5 orientation. apoptosis occurs.
DNA replication during the S phase requires a complex
orchestration of events; this discussion focuses on those events
terminate translation. The ribosome then dissociates, and the that are exploited for molecular diagnostic testing. Contained
peptide folds to its functional shape. within the double-stranded DNA helix are multiple origins of
replication. At each origin, the enzyme helicase disrupts and
DNA Replication and the Cell Cycle untwists the hydrogen bonds, separating the DNA strands and
After cells carry out their functions, they either divide via producing two replication forks. Here a deoxyribonucleotide
mitosis or die via apoptosis, also called programmed cell death. (deoxynucleotide triphosphate, or dNTP) polymerizes to form
468 PART V Leukocyte Disorders

B
Figure 32-9 A, The purine adenine forms two hydrogen bonds with the pyrimidine thymine. The purine guanine forms three hydrogen bonds with the
pyrimidine cytosine. B, The two strands maintain a consistent distance from each other, which allows DNA to twist into a helix.

new complementary strands (Figure 32-12). DNA replication replication origin, primase joins a primer to the 3 end of the
occurs bidirectionally from the two replication origin sites. 5-to-3 (top) template strand (Figure 32-13). Then DNA poly-
Each DNA strand in the replication fork serves as a template merase recognizes the free hydroxyl group on the 3 carbon of
for the formation of a daughter or complementary strand the last nucleotide in the primer and catalyzes the formation
through the activity of DNA polymerase.7 The DNA polymerase of phosphodiester bonds between the correct complementary
substrate is the free hydroxyl group located on the 3 carbon of nucleotide triphosphate and the primer, releasing the - and
a deoxyribonucleotide. DNA polymerase recognizes the group -phosphate groups. DNA polymerase continues adding deoxy-
and catalyzes the joining of the complementary deoxyribonu- ribonucleotides along the replication fork, going to the left of
cleotide. DNA is read 3 to 5 by DNA polymerase, and the the replication origin, producing the complementary strand
complementary strand is synthesized 5 to 3. called the leading strand.
A primer provides the free 3 hydroxyl group required for The second template strand, called the lagging strand, is also
DNA polymerase activity. Primers are short nucleotide poly- read in the 3-to-5 direction. To form a complementary strand,
mers complementary to the template. The hybridization of the a primer hybridizes to the exposed 3 end of the replication
primer to the template requires the enzyme primase. At the fork. To proceed in the 5-to-3 direction, nucleotides are added
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 469

Figure 32-10 RNA polymerase binds to a sequence of DNA called the promoter region, which causes the DNA strands to separate. Using one of the DNA
strands as a template, RNA polymerase moves along and simultaneously reads the DNA strand, forming the primary messenger RNA (mRNA) transcript by
joining the complementary ribonucleotides. The primary mRNA transcript consists of sequences called exons that provide coding information and introns that
are excised from mRNA. Introns may contain important information, and their functions remain under investigation.

Figure 32-11 The cell cycle consists of interphase and mitosis. Interphase is divided into G1, S, and G2 phases. Cell growth occurs during G1. During the
S phase, DNA synthesis (replication) occurs. The cell prepares for mitosis during the G2 phase. During mitosis the cell divides, producing two identical daughter
cells. Cells may also enter a quiescent phase called G0. During G0, the cell performs its function but does not replicate.
470 PART V Leukocyte Disorders

Figure 32-12 DNA replication occurs at replication origin sites throughout the DNA. Helicases separate the DNA strands, producing replication forks to
the left and right of the origins.

in fragments toward the origin of replication. As the left repli- lymphocytic leukemias (CLL),14-16 and 17% of acute lympho-
cation fork extends to open more of the template strands for blastic leukemias (ALL),17-19 are associated with a p53 mutation
replication, additional primers are hybridized, and DNA poly- or deletion (see Box 32-3). In summary, DNA synthesis and
merase uses the primers to initiate the formation of the com- accurate cell cycle control demand that the integrity of the
plementary strand, continuing until it meets a previously nucleotide sequence be maintained during DNA replication.
hybridized primer.
DNA polymerase not only joins nucleotides, it also degrades
MOLECULAR DIAGNOSTIC
the RNA primers and fills in the correct complementary deoxy-
TESTING OVERVIEW
ribonucleotides. Because the replication of the lagging strand
produces many small fragments, it is called discontinuous repli- DNA sequences are used to diagnose and monitor solid tumors,
cation, and the fragments are called Okazaki fragments. Finally, acute leukemia, myeloproliferative disorders, myelodysplastic
the enzyme ligase joins the discontinuous fragments. The rep- neoplasms, inherited thrombosis risk factors, and viral, para-
lication fork to the right (downstream) is replicated in the same sitic, and bacterial infections. Molecular diagnostic testing
fashion, although the lagging strand is now formed comple- exploits the enzymes and processes of DNA replication. Most
mentary to the top (5-to-3) strand, and the leading strand is molecular testing methods use replicationfor example, poly-
formed from the 3-to-5 strand; the opposite of the situation merase chain reaction (PCR)to make millions of amplicons
described occurs for the left replication fork (Figure 32-14). (copies) of a DNA sequence of interest. Further, creation of
The cell cycle is highly regulated. At certain critical points synthetic DNA allows the production of short sequences used
within the cycle, decisions are made to continue or begin cell as either primers or probes to locate specific DNA or RNA
death via apoptosis. This decision may depend on the state of sequences within vast populations of nucleic acids.
the DNA replicated (Figure 32-15). Normally, the cell detects Several specific mutations are associated with hematologic
errors made during replication and either corrects them or disease. These are detected by nucleic acid hybridization,
begins apoptosis. This prevents the persistence of daughter sequencing, or restriction fragment length polymorphism anal-
cells with genetic errors. If the sensing molecules fail, cell divi- ysis of amplified material. Messenger and ribosomal RNA also
sion may continue. Debilitating mutations that mediate cell may be amplified through a process called reverse transcriptase
cycle control may result in tumor formation. PCR (RT-PCR). Using mRNA, the existence of mutations that
One protein responsible for signaling damaged DNA is p53, are being actively translated can be detected. Assessment of
a tumor suppressor protein. Damaged cells with increased p53 mRNA shows whether a mutation is expressed in a certain cell
arrest cell division at G1, which allows time for DNA repair type or tissue and can be used to quantitatively determine the
(Figure 32-16). Cells with mutant p53 are unable to arrest level of transcription (number of copies) of a gene.
cells in G1; they continue the process of cell division with Most molecular tests use DNA amplification, generating
damaged DNA.8-10 If the cell can repair the DNA damage, the multiple amplicons of the target sequence. Amplification is
cell cycle continues. If the cell damage is too severe, the cell meant to be specific to the sequence of interest; however, it
undergoes apoptosis. Hematologic malignancies, such as 21% fails to differentiate among variant sources. Consequently, it is
of chronic myelogenous leukemias (CML),11-13 23% of chronic critical to eliminate contamination from previously amplified
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 471

B
Figure 32-13 A, Primases join primers to the single-stranded template strands. The primers must be oriented in such a way that the hydroxyl group on
the 3 end of the primers is available for deoxyribonucleotide addition by DNA polymerase. B, DNA polymerase extends the primer located on the 5-to-3
template strand, producing the complementary leading strand (blue). On the 3-to-5 template strand, DNA polymerase extends the primers, producing Okazaki
fragments. The primer ribonucleotides (red) are replaced with deoxynucleotides by DNA polymerase to produce the complementary lagging strand (green).

samples. Contamination can be avoided by designating sepa- mind, several techniques are presented and an example from
rate laboratory locations for each step and employing appro- hematopathology is given for each.
priate controls. Operators routinely employ ultraviolet (UV)
light and bleach to induce strand breaks in contaminating
NUCLEIC ACID ISOLATION (EXTRACTION)
DNA on work surfaces and a uracil-N-glycosylase system that
destroys previously amplified DNA. Isolating DNA from Clinical Specimens
In genetically based hematologic disease, mutations and Most molecular diagnostic tests begin with the isolation of
polymorphisms can occur that do not affect function. Indi- DNA or RNA from a patient sample. To test for a mutation in
viduals vary in genetic sequences coding for identical proteins. patient DNA, the patients DNA is isolated. To test for micro-
Such single nucleotide polymorphisms are commonly detected, organism DNA, as in an infection, DNA is also isolated from
but might not be associated with disease. With these caveats in the patient sample, because it will include the organism DNA.
472 PART V Leukocyte Disorders

Figure 32-14 Bidirectional DNA replication. The 5-to-3 parent strand serves as the template for producing the continuous leading strands on a replication
fork to the left of an origin. The 3-to-5 parent strand is the template for the lagging strands, which are produced in a discontinuous manner. The continuous
and discontinuous strands are reversed on the replication fork to the right.

molecular techniques, DNA must be isolated from the affected


tissues. Peripheral blood is adequate for infections with viruses
such as human immunodeficiency virus (HIV) and cytomega-
lovirus (CMV) that infect blood cells, whereas cerebrospinal
fluid is required for meningeal infections.
Whole blood is collected in an ethylenediaminetetraacetic
acid (EDTA) tube to prevent clotting and to inhibit enzymes
that may digest DNA. The white blood cells are separated and
ruptured using a solution of detergent and proteinase. A high-
salt solution removes the cellular debris and proteins, leaving
the DNA in the aqueous solution.
Next, the DNA is precipitated. DNAs phosphate backbone
is negatively charged, which prevents the DNA strands from
coming in close contact with one another, a prerequisite for
precipitation. The high salt level of the removal solution neu-
Figure 32-15 Toward the end of the G1 phase, the first critical point in tralizes the charges of the backbone, but the DNA is still soluble
the cell cycle is reached. At this critical point, the cell will either continue in the aqueous solution. The addition of isopropanol precipi-
into the S phase or go through apoptosis. The second critical point is located
tates the DNA, because nucleic acids are insoluble in alcohol.
at the end of the G2 phase. At this point, the cell will either continue into
The precipitated DNA appears as long, whitish strands in the
mitosis or initiate apoptosis.
solution. After washing with 70% ethanol and resuspension in
an aqueous buffer solution, the DNA is ready for molecular
The preferred nucleic acid for clinical diagnosis is DNA because testing.
it is inherently more stable than RNA and is less labor intensive If a delay in the molecular testing is necessary, the isolated
to isolate. DNA sample can be stored at 80 C indefinitely. Similar
Patient specimens for human DNA isolation include periph- procedures are used to extract DNA from dispersed cells from
eral blood, bone marrow, tissue biopsy specimens, needle aspi- bone marrow and needle aspirate specimens.20
rates, and cheek swabs. A blood specimen is appropriate for DNA from tissue suspected of being cancerous can be iso-
identifying an inherited defect. Every nucleated cell contains a lated from formalin-fixed, paraffin-embedded tissue sections
full complement of DNA. If an individual has inherited a mounted on glass microscope slides. Tissue is obtained from
mutation, it is present in the DNA of all their nucleated, non- the entire section or from a portion of the section by microdis-
gamete (somatic) cells. The DNA of nucleated white blood cells section, either by scraping or by laser. The tissue is degraded
reveals inherited mutations. In solid tumors, somatic (acquired) by an enzyme called proteinase K to break open the cells and
mutations are detected by analyzing DNA from the suspect release the DNA. The sample is then heated to 94 C for several
tissue. For identification of infectious disease organisms by minutes to inactivate the proteinase K and to degrade other
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 473

Figure 32-16 A, Cells with no DNA damage and normal levels of p53 divide normally. B, Damaged DNA within cells causes an increased concentration
of p53. The physiologic increase in p53 causes the cell to arrest in G1, which allows for cellular repair. If the cell is too damaged for repair, it will go through
apoptosis. C, Cells with damaged DNA and mutated or nonfunctioning p53 cannot arrest in G1 for cell repair. These cells either go through cell death via
necrosis or continue cell division, beginning the process of tumor formation.

proteins, and the DNA is purified and precipitated as previ- mRNA. The mRNA does not represent all the information
ously discussed.21 In addition to paraffin-embedded samples, stored in the DNA, only those genes being expressed. Conse-
fresh or frozen tissue samples are appropriate for DNA quently, mRNA provides quantitative information on the genes
isolation. Quickly thawing and mincing the frozen tissue pre- being expressed in a cell at the time the specimen is collected.
pares the sample for DNA isolation. The minced tissue is mixed The steps of RNA isolation are (1) RNA release by cell lysis,
with an extraction buffer to release the DNA from the cells; it (2) RNase inhibition with strong chemical agents such as urea
is then purified and precipitated as described earlier. or guanidine isothiocyanate, (3) protein and DNA removal,
and (4) RNA precipitation. In step 3, extraction is performed
Isolating RNA from Clinical Specimens using phenol at a pH of 4, chloroform, and isoamyl alcohol.
RNA isolation poses greater technical challenges than DNA These separate the DNA and protein into the organic phase,
isolation. Ubiquitous ribonucleases (RNases) degrade RNA. while the RNA remains in the aqueous phase. RNA resists
These enzymes are the bodys primary defense against patho- acidic pH, whereas DNA is readily depurinated, because acid
gens and are found on mammalian epidermal surfaces; cleaves the bond between the purine base and the deoxyribose
therefore, they contaminate all laboratory surfaces.22 Clinical sugar. Therefore, acidic phenol preferentially isolates and
laboratories that isolate RNA must be RNase free, which neces- preserves RNA while the genomic DNA (all the DNA) is
sitates costly precautions and decontamination steps.23 partitioned along with contaminating proteins, lipids, and car-
The isolated RNA includes mRNA, rRNA and tRNA, all of bohydrates. As with DNA, precipitating the RNA from the
which participate in protein synthesis. Depending on cell type, aqueous phase requires the addition of salt to neutralize the
mRNA may comprise only 3% to 5% of the total cellular RNA; charge of the phosphodiester backbone and ethanol to make
therefore, a large specimen may be needed to obtain adequate the nucleic acid insoluble.24,25
474 PART V Leukocyte Disorders

the first complementary deoxyribonucleotide to the primer.


AMPLIFICATION OF NUCLEIC ACIDS
The polymerase continues down the template strand at 1000
Polymerase Chain Reaction for nucleotides per second, extending the complementary strand
Amplifying DNA to eventually produce a complete daughter strand that contin-
PCR is the principal technique in the clinical molecular labora- ues to the 3 end of the template.28 This completes one PCR
tory. PCR is an enzyme-based method for amplifying a target cycle. In the second cycle, the temperature changes are repeated,
sequence to allow its detection from a small volume of mate- and the first-cycle product becomes the template for a daughter
rial.26 Sickle cell anemia results from a single -globin nucleo- strand. After the second cycle, the daughter strand is bounded
tide substitution (point mutation) in which an adenine replaces by the primer sequences at the 5 and 3 ends, producing a
a thymine. Detecting this mutation from among 6 billion fragment of DNA of the desired length. In 25 to 40 subsequent
nucleotides would be like finding a needle in a haystack if only cycles, this DNA of specific length and sequence, called an
a few cells were assessed. When millions of -globin copies are amplicon, is reproduced millions of times.29,30
produced, however, the mutation is easily detected. Primer annealing accounts for PCR specificity, and primer
As with natural DNA replication, PCR amplification requires design is crucial for achieving confidence in data analysis.
primers. In testing for the sickle cell mutation, for example, When testing for a mutation, the primer set must flank the
selected primers flank (i.e., bind on either side of) the -globin target region without annealing to other regions in the sample.
gene sequence containing the mutation. The total base pair When testing for sequences from a specific disease-causing
(bp) length of the primer sequences plus the target sequence microorganism, the primers must anneal to the target region
can vary, but is 110bp for the -globin gene, a typical sequence of that organisms DNA, but not to the hosts DNA or the DNA
length for many mutation sites (Figure 32-17).27 Besides of other organisms. Wherever primers anneal, whether by
primers, the PCR master mix reagents include a heat-insensitive intention or not, they form starting points for extension.
DNA polymerase called Taq polymerase, isolated from the Commercial kits contain primer sets that have been tested
thermophilic bacterium Thermus aquaticus, and the deoxyribo- for annealing specificity, but care must be taken to use the
nucleotides deoxyadenosine triphosphate (dATP), deoxythy- optimal annealing temperature. Even if the primer is properly
midine triphosphate (dTTP), deoxyguanosine triphosphate designed, it can anneal to nonidentical regions if the annealing
(dGTP), and deoxycytidine triphosphate (dCTP) in a magne- temperature is too low. Laboratory researchers who design
sium buffer. primers look to digital algorithms such as the Basic Local
The DNA is first denatured at 95C, which separates the Alignment Sequence Tool (BLAST).31 Further, complemen
strands; then cooled to the primer annealing temperature of 40 tarities must be avoided between the primers themselves to
to 60C; then warmed to 72C to promote Taq polymerase prevent hybridization to one another, which forms undesirable
promoted chain extension, in which nucleotides are added to primer dimers.
the primers (Figure 32-18). The annealing temperature is Controls are essential. The three controls required for PCR
optimized for each set of primers. A thermocycler is used are the negative, positive, and no-DNA controls. All three are
to accurately produce and monitor the rapid temperature included in each run. The negative control consists of DNA
changes. known to lack the sequence of interest; the positive control
Once the double-stranded DNA is denatured, one primer contains the target sequence. Comparison of the bands in the
anneals to the 5-to-3 strand and the other to the 3-to-5 patient specimen electrophoretic lanes to bands in the negative
strand. Both primers possess a free 3 hydroxyl group. The Taq and positive control lanes determines whether the target DNA
polymerase recognizes this hydroxyl group, reads the template, sequence is present in the patients DNA. The no-DNA control
and catalyzes formation of the phosphodiester bond joining detects master mix contamination. A band in the no-DNA lane

Target -globin DNA

Primer extension

Figure 32-17 Flanking primers PCO3 and PCO4 are used to amplify the target -globin DNA. One primer (PCO3) joins with the 3 end of the 5-to-3 DNA
strand. The other primer (PCO4) anneals to the 3 end of the 3-to-5 DNA strand. These primers form the site for extension during polymerase chain
reaction.
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 475

Target -globin DNA

Taq polymerase adds complementary deoxyribonucleotides,


making dsDNA

Target -globin DNA amplified

Figure 32-18 Application of polymerase chain reaction (PCR) to target -globin DNA. PCR amplifies the target DNA, making millions of copies of the target
DNA after 30 cycles. dsDNA, double-stranded DNA; ssDNA, single-stranded DNA.
476 PART V Leukocyte Disorders

Figure 32-19 The BCR gene is present on chromosome 22 and the ABL gene is located on chromosome 9. The Ph chromosome results from the trans
location of the ABL gene to chromosome 22, which places the ABL gene next to the BCR gene and produces a chimeric BCR/ABL gene. The transcription of
the BCR/ABL gene produces a chimeric messenger RNA (mRNA) consisting of a portion of the BCR gene and a portion of the ABL gene.

indicates DNA contamination, which renders the entire test mRNA. Reverse transcriptase recognizes the hydroxyl group on
result unreliable.32 the last nucleotide of the primer and reads the mRNA template
strand, then adds the correct complementary deoxyribonucleo-
Reverse Transcription Polymerase Chain tide. Reverse transcriptase continues up the mRNA template
Reaction for Amplifying RNA strand, joining the complementary deoxyribonucleotides to
Some hematology analyses require mRNA. Genetically altered the growing cDNA strand to form the mRNA-cDNA hybrid.
mRNA sequences translate to an altered protein. For instance, Subsequently, heat denaturation breaks the hydrogen bonds
the Philadelphia chromosome (Ph), carrying the mutation between the mRNA-cDNA hybrid, separating the two strands.
t(9;22)(q34;q11.2), is present in 95% of CML cases plus 20% The cDNA strand then acts as a template for replication by
of adult ALL and 5% of pediatric ALL cases, and in rare instances DNA polymerase. In the next step, the single-stranded cDNA
in acute myeloid leukemia.33,34 Ph results from a reciprocal is amplified as in DNA-based PCR using primers specific for a
translocation of the ABL (Ableson) gene on chromosome 9 to target sequence in the BCR gene and ABL gene. DNA poly-
the breakpoint cluster region (BCR) of chromosome 22, pro- merase extends the primers, forming a double-stranded cDNA
ducing a BCR/ABL hybrid (Figure 32-19).35-37 Transcription of of the target chimeric gene. The cycling continues, resulting in
BCR/ABL produces a chimeric mRNA made up of fragments millions of copies of the BCR/ABL sequence.39,40
from both the BCR and ABL genes. Translation generates a
fusion protein, tyrosine kinase, that alters normal cell cycle
DETECTION OF AMPLIFIED DNA
control, which results in unrestrained cell proliferation.38
RT-PCR is performed to detect the chimeric mRNA, thus the Amplified target DNA may be detected by gel electrophoresis
mRNA template is preferred to DNA. Although the mutation using ethidium bromide (EtBr) or SYBR green fluorescent dyes,
is present at the DNA level, the position at which the two or autoradiography for visualization; by restriction enzyme
chromosome sections join is variable, whereas the chimeric action followed by gel electrophoresis; or by hybridization to
mRNA is always the same. Also, the DNA includes untranslated a known sequence (probe).
introns, which make the chimera too long to replicate. Physi-
ologic mRNA excision and splicing yields a much shorter target Gel Electrophoresis
that is more easily amplified. Nucleic acid phosphate groups confer a net negative charge.
In RT-PCR, the reverse transcriptase enzyme produces comple- Consequently, in electrophoresis, the rate at which DNA frag-
mentary DNA (cDNA) from mRNA present in a total RNA ments (amplicons) pass through gels is proportional to their
sample extracted from patient blood cells (Figure 32-20). PCR mass only and, unlike proteins, not their relative charge. DNA
subsequently amplifies the cDNA. The RT-PCR master mix fragment mass is a function of the length in base pairs (bp) or
includes an oligo(dT), random, or specific primer; reverse tran- kilobase pairs (kb, 1000 bp). Fragments are sieved through an
scriptase; deoxyribonucleotides; primers; the mRNA template; agarose or polyacrylamide gel matrix by passing a current through
and Taq polymerase. the gel as it is bathed in a buffered conducting salt solution.
The first step employs reverse transcriptase and a specialized Electrophoresis gel pore diameter is a function of gel concentra-
primer to produce an RNA-cDNA hybrid. The primer, called tion. The pores of an agarose gel are larger than the pores of a
oligo(dT), is a series of thymine nucleotides. Most mRNAs polyacrylamide gel. When larger fragments (500bp to 50kb)
possess a string of adenine nucleotides on the 3 end called the are to be separated, an agarose gel is most effective. For smaller
polyA tail. The oligo(dT) primer anneals to the polyA tail of DNA fragments (5 to 1000bp), a polyacrylamide gel is used.41
Figure 32-20 Reverse transcriptase
DNA polymerase extends the ABL primer
polymerase chain reaction (RT-PCR)
produces complementary DNA (cDNA)
from messenger RNA (mRNA). This
diagram shows the RT-PCR steps used
to produce amplified BCR/ABL cDNA.
Initially an oligo(dT) primer (consisting of
a series of deoxythymidine nucleotides)
anneals to the 3-polyA tail of the
chimeric BCR/ABL mRNA. Reverse
transcriptase elongates the primer,
producing an mRNA-cDNA hybrid. Heat
denaturation breaks the hydrogen bonds
holding the hybrid molecule together,
releasing the single-stranded (ss) BCR/
ABL cDNA. Next, a primer specific for
the ABL gene is annealed to the cDNA.
DNA polymerase extends primers DNA polymerase elongates the primer,
producing the double-stranded (ds)
BCR/ABL cDNA. The cDNA becomes
single stranded by heat denaturation.
Then the ABL primer as well as a primer
specific for the BCR gene anneal to the
ss cDNA. DNA polymerase elongates the
primers, producing ds BCR/ABL cDNA.
The cycle is repeated 20 to 40 times,
producing millions of copies of the ds
BCR/ABL cDNA.
478 PART V Leukocyte Disorders

Figure 32-21 Gel electrophoresis pattern of a DNA sample. A, Molecular


size marker (ladder); B, positive control; C, negative control; D, no-DNA
control. By comparing the bands present in the gel with the molecular size
markers, the mass of each band, measured in base pairs, is determined. For
example, in the positive control sample, the three bands are 184, 110, and
89bp. Positive, negative, and no-DNA controls must be used when perform
ing gel electrophoresis. The positive control contains the target DNA
sequence, and the negative control lacks this sequence. The no-DNA control
sample lacks DNA. No banding should be present in the no-DNA control. If
bands are present, contamination of samples occurred during the testing
process.

Figure 32-22 Autoradiograph of amplified DNA illustrating a sample


DNA fragments (amplicons), controls, and a mass marker
positive for the sequence of interest.
or ladder are pipetted into the sample wells near the anode.
Electrical current moves the negatively charged fragments
toward the positive electrode. Smaller fragments move faster alternatively, probes may be hybridized with enzyme-conjugated
(farther) than larger fragments. The ladder, composed of frag- nucleotides. The enzymes most commonly used are horseradish
ments of known masses, measured in base pairs or kilobase peroxidase (HRP) and alkaline phosphatase (AP), which cleave
pairs, runs alongside the sample and control lanes. Amplicon luminol or other synthetic chemiluminescent substrates to
masses of the sample and control are determined by comparing release visible light. Light generation is confined to those bands
their bands with the bands of the ladder (Figure 32-21). that contain HRP- or AP-conjugated DNAs.
EtBr, SYBR green, or autoradiography may be employed to Gel electrophoresis is appropriate when the goal is to deter-
detect the controls, size marker, and patient DNA fragments. mine qualitative target presence or absence. Typical target
EtBr, a hydrophobic molecule about the same mass as a purine mutations in hematology are four that raise thrombosis
or pyrimidine base, becomes intercalated between the bases of risk: the coagulation factor V Leiden mutation, prothrombin
a DNA double helix or between the bases of a nucleic acid G20210A, MTHFR C677T, and MTHFR A1298T. Three others
fragment, rendering the nucleic acid fluorescent when exposed are mutations that affect the metabolism of the antithrom-
to UV light. EtBr may be pipetted directly into the gel as it is botic drug warfarin. These three, CYP2C9*2, CYP2C9*3, and
prepared or diluted in a buffer and applied after electrophore- VKORC1, are among the first targets of pharmacogenetic
sis. UV light causes EtBr to fluoresce orange, illuminating the testing. Patients positive for the presence of any one or more
nucleic acid bands. EtBr is a mutagen, so many laboratories of these mutations are sensitive to warfarin and require smaller
substitute the safer SYBR green. SYBR green binds to the minor than the usual dosages. A normal warfarin dosage risks causing
groove of nucleic acid helices and fluoresces. hemorrhage in warfarin-sensitive patients. Several of these
For autoradiography, deoxyribonucleotides that are incor- example mutations may also be identified using restriction
porated during the PCR elongation step, usually the adenine endonuclease methods.
nucleotide, are conjugated to a radioactive -phosphate group
([32P]dATP). After electrophoresis, x-ray film is placed over Restriction Endonuclease Methods
the dried gel. The radioactivity in the amplified DNA fragments One method to determine whether an amplified target DNA
exposes the film, producing a banding pattern that is inter- fragment contains a mutation of interest uses enzymes called
preted by the laboratory scientist (Figure 32-22).42 restriction endonucleases (also known as restriction enzymes).
An enzyme-based approach may be used in place of auto These enzymes are produced naturally in bacteria and are so
radiography, thereby eliminating the health, storage, and named because they restrict foreign (phage) DNA from enter-
disposal concerns associated with radioisotopes. Enzyme- ing and destroying the bacterium. Each restriction enzyme rec-
conjugated nucleotides are incorporated during extension; ognizes a specific nucleotide sequence and cuts both strands of
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 479

and produces four bands of lengths 37, 82, 104, and 141bp
(see Figure 32-1).

Nucleic Acid Hybridization and


Southern Blotting
Another detection method employs a nucleic acid probe
designed to hybridize (base-pair) to a selected sequence.
Nucleic acid hybridization may be combined with restriction
enzyme digestion to make identifications on an electro
phoresis gel.47,48
Dr. Edwin Southern developed the classic Southern blot
hybridization, which was originally performed using isolated
DNA without amplification, but is now performed using PCR-
amplified DNA. The Southern blot procedure, which takes 3
days to perform, is now confined to research applications, but
pointed the way to RFLP and probe hybridization technology.
Restriction endonuclease EcoRI cuts DNA at many sites (Figure
32-25). Gel electrophoresis separates the fragments. The sample
Figure 32-23 Restriction enzymes recognize a specific nucleotide
is then acid-depurinated to nick the fragments. Sodium
sequence in DNA called the restriction site and cut both strands of the DNA.
hydroxide next denatures the DNA, producing single-stranded
A, The scissors represent a restriction enzyme recognizing one restriction
site in the amplified DNA, producing two restriction fragments. B, Two restric DNA without changing its nucleotide sequence or two-
tion sites are present in the amplified DNA. After the restriction enzyme cuts dimensional position on the gel. The single-stranded DNA is
the DNA, three restriction fragments are present. transferred to a nitrocellulose filter by electric current or capil-
lary action, so that the nitrocellulose filter reflects the banding
pattern. The DNA is permanently affixed to the nitrocellulose
the target DNA at the sequence, producing restriction fragments. filter by baking and UV cross-linking.49,50
Recognition sequences can be 4 to 15 nucleotides long. There are In the classic Southern blot procedure, detection of the
hundreds of commercially available restriction endonucleases, band containing the sequence of interest requires a radioactive
which allows recognition of many sequences. The number of or enzyme (horseradish peroxidase or alkaline phosphatase)
restriction fragments produced depends on the number of conjugated, single-stranded probe complementary to the target
restriction sites present in the amplified target.43,44 Enzyme sequence. The probe hybridizes to the target DNA, leftover
action at one restriction site produces two restriction frag- unhybridized probe is washed off, and the hybridized bands
ments, action at two restriction sites produce three restriction are visualized by autoradiography. Southern blot analysis may
fragments, and so on (Figure 32-23). A restriction enzyme also be performed using a fluorescently labeled probe instead
detects even a single base substitution, because the mutation of a radioactive one.51,52
alters its sequence and prevents digestion at the site. A time-honored application of the Southern blot technique
A restriction fragment length polymorphism (RFLP) is a muta- is detection of B-cell immunoglobulin heavy chain gene or
tion or polymorphism-induced change in the position of the T-cell receptor (TCR) gene rearrangement. Each immunoglobu-
restriction fragment. The factor V Leiden mutation is an excel- lin heavy chain or TCR gene possesses a unique sequence, the
lent example of RFLP. Individuals possessing this mutation result of somatic rearrangement. Rearrangement joins distinct
have an increased risk of venous thrombosis. The factor V gene segments from pooled clusters with unique sequences.
Leiden mutation results from the replacement of guanine with Thus, each B cell and its progeny produce a specific antibody,
adenine at position 1691 (G1691A) of the coagulation factor and each T cell and its progeny possess a specific cell receptor.53-55
V gene.45,46 The mutation alters a site normally detected and Lymphoproliferative disorders arise from a malignant transfor-
cut by the restriction enzyme Mnl1. The wild-type (normal) mation of a B or T cell. The malignant cells, no longer under
factor V amplicon is 223bp long with two Mnl1-specific sites. the control of cell division regulatory proteins, divide uncon-
After PCR and incubation with Mnl1, the wild-type amplicon trollably, forming a malignant clone. All the cells within the
is cut to three restriction fragments, separable using polyacryl- clone contain the same gene rearrangement; thus the cells are
amide gel (Figure 32-24). The fragments are 37, 82, and 104bp monoclonal. Gene rearrangement analysis detects the mono-
long. The mutant gene generates only two fragments with clonal population and determines whether it is a B-cell or T-cell
lengths of 82 and 141bp. A sample from an individual homo- lymphoproliferative disorder.
zygous for the wild-type gene generates the three anticipate The appropriate specimens for gene rearrangement analysis
fragments; 37, 82, and 104bp. A sample from an individual are bone marrow, blood, or tissue from the tumor site. After
homozygous for the factor V Leiden mutation possesses two DNA extraction, restriction enzymes EcoRI, BamHI, and HindIII
copies of the mutated factor V gene and generates only two cut the DNA into restriction fragments. Agarose gel electropho-
bands, 82 and 141bp. A sample from a heterozygous indi- resis separates restriction fragments. The bands are then depu-
vidual possesses one normal and one mutated factor V gene rinated, denatured, and transferred to a nitrocellulose filter.
480 PART V Leukocyte Disorders

GGAG

GGAG 3
5
CCTC
3 5
CCTC

GGAG
AGAG 3
5
CCTC
3 5
TCTC

GGAG
GGAG
3
5

GGAG
AGAG
3
5
Figure 32-24 The restriction site GGAG is recognized by the restriction enzyme Mnl1. The normal coagulation factor V gene contains two restriction sites
for Mnl1. In the factor V Leiden mutation gene, the substitution of an A for a G destroys one of the restriction sites for Mnl1. Thus, the mutated factor V gene
possesses only one restriction site for Mnl1. A, The amplified target sequence for the factor V gene in a normal individual contains two restriction sites for
Mnl1, so that restriction fragments with the sizes of 104, 82, and 37bp are produced. B, In an individual who is homozygous for the factor V mutation there
is only one restriction site for Mnl1, so that two restriction fragments of the sizes 141 and 82bp are produced. C, An individual who is heterozygous for the
factor V mutation has a normal and a mutated factor V gene. Mnl1 produces four restriction fragments of 141, 104, 82, and 37bp.

Probes hybridize the nucleotide sequences in either B- or T-cell be visualized. For health and disposal reasons, the use of radio-
genes, and autoradiography visualizes the banding pattern. The active isotopes is restricted in the clinical setting, but alterna-
presence of a distinct band represents a monoclonal cell popu- tive labeling techniques and detection kits are commercially
lation. A polyclonal population, consisting of cells with differ- available. For instance, fluorogenic acridinium ester molecules
ent gene rearrangements, appears as a smear with no distinct may be incorporated as a direct label. Sodium tetraborate solu-
banding pattern.56-58 Gene rearrangement analysis provides tion, containing 1% Triton X-100, degrades acridinium ester
physicians with an important tool to diagnose and monitor the on unhybridized probes, whereas base pairing of the probe to
progress of lymphoproliferative disorders. its target protects the ester.59 The bound ester is detected by a
brief emission of light after addition of hydrogen peroxide.
Hybridization Labels Biotin and digoxigenin are two commonly used indirect
Labels are visualizing molecules conjugated to, or incorporated labels. Biotin-labeled probes bind avidin. Avidin is in turn
into, probes or primers. Radioactivity is incorporated by intro- linked to a fluorescent label or to a detectable enzyme (see
ducing isotope-labeled nucleotides during probe synthesis. Figure 32-26). Digoxigenin-labeled probes are bound by an
Radioactivity is detected by applying the sample to photo- antidigoxigenin antibody conjugated to a fluorescent molecule
graphic film, which is an example of the use of a direct label or enzyme. Alkaline phosphatase or horseradish peroxidase are
(Figure 32-26). Direct labeling means that the molecule can be the enzymes most commonly used in indirect labeling systems.
visualized without the addition of a reporter system, whereas Their activity cleaves substrates to produce visible pigments or
indirect labels require additional molecules and reactions to chemiluminesence.60-62
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 481

Nucleic acid

Label Indirect Direct


method

Biotin Digoxygenin Radioactive Fluorogenic


Acridinium ester

Bound to Avidin Antidigoxigenin

Label Enzymatic Fluorogenic

Peroxidase Phosphatase

Chromogenic DAB NBT


substrates TMB BCIP

Chemiluminescent
substrates Diacylhydrazides Dioxetanes

Figure 32-26 To detect the presence of target sequences in DNA


samples, nucleic acids are labeled directly using a radioactive isotope, acri
dinium ester, or a fluorophore. Alternatively, they are labeled indirectly with
biotin and digoxigenin, which are detected using avidin or antidigoxigenin,
respectively, and chromogenic or fluorogenic substrates. Chromogenic sub
strates for peroxidase and phosphatase include 3,3-diaminobenzidine
(DAB), 3,3,5,5-tetramethylbenzidine (TMB), nitroblue tetrazolium (NBT), and
5-bromo-4-chloro-3-indoylphosphate (BCIP).

dideoxyguanine triphosphate, and dideoxythymine triphos-


Figure 32-25 Southern blot steps. 1, DNA is cut with the restriction phate each are included in the PCR master mix, over a number
endonuclease EcoRI, which produces many restriction fragments. 2, DNA of cycles a series of fragments that terminate at each successive
fragments are separated on an agarose gel. 3, DNA fragments are trans base is produced. This is called a nested series of fragments. The
ferred to a nitrocellulose filter. 4, A radioactive probe is hybridized to the DNA dideoxynucleotides are fluorescently labeled to produce a differ-
fragments on the filter. 5, Autoradiography is used to visualize the hybridized ent color for each base, and the fragments are subjected to capil-
DNA probe, the detection of which indicates the presence of the given lary electrophoresis. The fragments with their labels pass through
sequence in the sample. the beam of a detecting laser one by one, with their order based
on their length, which allows their sequence to be read. Because
DNA Sequencing DNA is double-stranded, two PCR primers would produce two
The ability to read the sequence of the nucleic acid has been just series of nested fragments and the detector would read two bases
as important as PCR in the development of molecular biology.63 at each position. To avoid this, the PCR reaction is done with
A combination of these two important techniques (cycle one primer, which produces single-sided PCR.
sequencing) has made DNA sequencing more efficient and its With the sickle cell mutation, for example, sequencing of
analysis less subjective. In cycle sequencing, the order of the either, but not both, strands will show whether the mutation
nucleotide bases is determined after amplification.64 Cycle (adenine to thymine) is present. Each cell has two copies
sequencing is applied in molecular testing to assess amplified (alleles) of somatic genes; therefore, sequencing will produce
sequences for insertions, deletions, or mutations, such as coag- a nested series of fragments from each allele. If the patient is
ulation factor V Leiden mutation or -globin mutation. homozygotic, the two nested series of fragments will be identi-
Cycle sequencing is based on dideoxynucleotide terminator cal, whether wild-type or mutant. If the patient is heterozy-
sequencing.65 The addition of nucleotides to a growing polymer gotic, both wild-type and mutant fragments will be produced
requires a 3 hydroxyl group on the last added nucleotide and a in the single-sided PCR, generating two nested series of frag-
triphosphate group on the 5 end of the next nucleotide to be ments. In analysis of this sequence, both adenine and thymine
added. If a nucleotide lacks the 3 hydroxyl group, it can be signals will be present at the position of the mutation, but
incorporated but cannot be added to, so the fragment terminates because only half the templates contain each sequence, the
at the defective base (Figure 32-27). If small concentrations of signals of the adenine and thymine will be half as strong.
dideoxyadenosine triphosphate, dideoxycytosine triphosphate, Sequencing is a reliable method for detection of mutations or
482 PART V Leukocyte Disorders

5 5 5
Primer Primer Primer
G A C T AOH G A C T AOH G A C T AOH

C T G A T G T A C C T G A T G T A C C T G A T G T A C

5 5 5
G A C T A CH G A C T A COH G A C T A COH

C T G A T G T A C C T G A T G T A C C T G A T G T A C

5 5
G A C T A C AH G A C T A C AH

C T G A T G T A C C T G A T G T A C

5
G A C T A C A TH

C T G A T G T A C

Figure 32-27 Gene sequencing technique. Cycle sequencing of a DNA template produces a nested series of fragments that differ by one nucleotide each.
The template is amplified by polymerase chain reaction (PCR) using a single primer sequence (single-sided PCR). The PCR master mix includes small amounts
of dideoxynucleotides that are fluorescently labeled, with each of the four bases conjugated to a different fluorophore. When a dideoxynucleotide is incorporated
into the growing polymer, extension ceases.

single nucleotide polymorphisms in DNA, but it is expensive The key to real-time quantitative PCR is fluorescence reso-
and requires significant instrumentation, so it is not often used nance energy transfer (FRET).69 Probes or primers are labeled
in the clinical setting. with fluorophores designed to fluoresce only upon binding
their selected nucleic acid sequences. The unbound fluoro-
phore possesses two active sites: a fluorescing reporter site and
REAL-TIME POLYMERASE CHAIN REACTION
a quencher site separated by the selected base sequence. When
In contrast to standard or end-point PCR, real-time quantitative unbound, the quencher site draws energy from the reporter site
PCR measures the change in nucleic acid amplification as rep- and thereby extinguishes its fluorescence. Upon binding, the
lication progresses using fluorescent marker dyes.66 The time quencher site becomes physically removed from the vicinity of
interval, expressed as the number of replication cycles, required the reporter, which permits the reporter site to fluoresce. As
to reach a selected fluorescence threshold is proportional to the cycling proceeds and more amplicons are produced, the fluo-
copy number of target molecules in the extracted sample.67 rescent signal grows until it exceeds the threshold. SYBR green,
Typical targets in this technique include viruses, bacteria, and which nonspecifically binds the minor groove of double-
tumor cells with discernible somatic mutations. Real-time ther- stranded DNA, exhibits FRET. TaqMan (Applied Biosystems,
mocyclers, such as the LightCycler (Roche Applied Science, Carlsbad, CA) is the prototype FRET-based specific probe, and
Indianapolis, IN), first developed in 1993, record the dynamic scores of patented competitors, exploiting a variety of creative
(delta) rise in fluorescence intensity during PCR. Real-time physical properties, are available to the operator.70
thermocyclers assay multiple samples from various sources in
replicate concurrently with internal standards to achieve a Minimal Residual Disease in Leukemia
clinical accuracy hitherto unattainable using phenotypic or cul- Real-time quantitative PCR provides the opportunity to
tural assay techniques.68 measure minimal residual disease in leukemia, a key indicator of
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 483

treatment efficacy, clinical remission, and prognosis.71,72 Cur- swabs, anaerobes from wound swabs, and bacteria from urine
rently, chemotherapy, radiation therapy, and peripheral blood or other body fluids can be detected within hours of collection.
stem cell transplantation reduce leukemic cells to levels unde- Antibacterial therapy can be initiated based upon the speedy
tectable first by visual bone marrow smear or peripheral blood results of molecular susceptibility testing. Real-time quantita-
film review and later by flow cytometry assay.73 This is called tive PCR is the reference method for detection and quantifica-
minimal residual disease, but despite the seeming disappearance tion of methicillin-resistant Staphylococcus aureus (MRSA),
of leukemic cells as recorded by these phenotypic assays, 1010 vancomycin-resistant enterococcus, and opportunistic Clos-
malignant cells may continue to reside undetected in blood, tridium difficile. Molecular diagnostic techniques are effecting
marrow, or lymphatic tissue.74 Real-time quantitative PCR in identifying and monitoring malarial and other blood-borne
identifies residual cells and helps guide the types and intensity parasites. The challenge to primer and probe developers is to
of therapy with the goal of molecular remission. Subsequent select sequences that are specific enough to avoid false positives
to remission, periodic real-time quantitative PCR assays may caused by nonpathogenic strains, sensitive enough to posi-
be used to detect early relapse and drug resistance, enabling tively identify infectious strains, and flexible enough to remain
the hematologist to initiate follow-up therapy.75 effective as pathogenic microorganisms mutate and evolve.
Real-time quantitative PCR may detect a single malignant Clinical relevance is important when assessing infectious
cell within a population of a million cells, providing unparal- disease using molecular techniques. Such methods allow mil-
leled sensitivity. Manufacturers compete to develop series of lions of copies to be generated from a single DNA sequence
FRET-labeled probes that are comprehensive, speedy, and spe- from a microorganism or virus. Theoretically, the presence of
cific for various hematologic diseases.76 Current applications a single organism can lead to a positive test result, but a single
include detection of BCR/ABL (Ph) in CML and some acute organism may not be clinically relevant when monitoring viral
leukemias (see Figure 31-19); JAK2 (just another kinase or loads. Standard curves of template number are crucial to data
Janus kinase) in the myeloproliferative neoplasms polycythemia interpretation. Also, because DNA survives the organism, a
vera and essential thrombocythemia; the t(15;17)(q22;q21) muta- positive result on a test for a given sequence does not guarantee
tion in acute promyelocytic leukemia; and gene rearrangement in that the organism was viable at the time of sampling.
lymphatic neoplasms.77
Real-time quantitative PCR may also exploit gene rearrange-
CURRENT DEVELOPMENTS
ment in lymphomas or lymphocytic leukemias to test for
minimal residual disease. Because the rearranged base sequence DNA amplification, hybridization, characterization, and
varies depending on the individual patients malignant clone, sequencing methods continue to be automated and miniatur-
the patients blood must first be collected and the tumor cell ized, providing ever greater sensitivity and reliability coupled
DNA cloned and sequenced.78 Thereafter, primers and probes with short turnaround time and technical simplification. Auto-
may be designed (and labeled with SYBR green or FRET probes) mation reduces pipetting errors and improves throughput. Gel
to replicate and detect the patients sequence.79 This process is analysis employs automated fluorescence readers with internal
called allele-specific oligonucleotide PCR. gel standards. Microarrays are silicon, plastic, or glass micro-
Although no purpose is served by quantitative measure- chips that resemble computer chips but are imprinted with
ment, the specificity and speed of real-time PCR mean that it nucleic acid probes from DNA, cDNA, and mRNA, and provide
is effective for the qualitative detection of germ-cell mutations the opportunity to detect hundreds of polymorphisms within
such as the factor V Leiden mutation, prothrombin G20210A, minutes. Nanotechnology permits the analysis of biologic
MTHFR C677T, MTHFR A1298T CYP2C9*2, CYP2C9*3, and molecules less than 100nm long, and the technologies of
VKORC1. genomics are being extended to proteomics, the molecular
analysis of proteins, and enzynomics, the analysis of enzymes.
Infectious Disease Load There is a demand for medical laboratory scientists with both
Real-time quantitative PCR can detect and quantitate a number technical and interpretive expertise. Molecular diagnostics is
of blood-borne viruses: hepatitis B and C viruses, human papil- currently the most rapidly growing segment of medical labora-
lomavirus, CMV, Epstein-Barr virus, and HIV.80 Human bacte- tory science and promises to revolutionize laboratory tech-
rial pathogens such as -hemolytic streptococcus from throat niques in all disciplines.

SUMMARY
DNA directs cell function as described by the central dogma. The mRNA transports code from the nucleus to the cytoplasm,
DNA retains the genetic code and reproduces itself through where it is translated in cytoplasmic ribosomes.
replication. Translation depends upon tRNA, small RNA molecules designed
The genetic code is transcribed from DNA to mRNA. to transport and add amino acid to growing peptide chains in
During transcription, untranscribed DNA introns are excised. cytoplasmic ribosomes.
484 PART V Leukocyte Disorders

DNA consists of a five-carbon sugar (deoxyribose), a phosphate Four areas of hematopathologic molecular diagnostic testing
group, and a nitrogenous base. The bases are either purines or include:
pyrimidines. Detection of somatic mutations in hematologic malignancies
The purines in DNA are adenine (A) and guanine (G) Detection of inherited hematologic and thrombotic disorders
The pyrimidines in DNA are thymine (T) and cytosine (C). Detection of hematologically important pathogens
DNA is a double-stranded molecule held together by hydrogen Monitoring of cancer therapies
bonding between the bases, A:T and G:C. Blood, bone marrow, tissue biopsy samples, and fine-needle aspi-
DNA is denatured by heat, which breaks the hydrogen bonds to rates are specimens used for DNA and RNA isolation.
produce single-stranded DNA. DNA is amplified by in vitro PCR and RT-PCR.
DNA strands are antiparallel. Amplified DNA is detected by:
The tumor suppressor protein p53 detects damaged DNA and either Gel electrophoresis
halts cell division for DNA repair or begins the process of apoptosis. Restriction endonucleases
RNA is a single-stranded molecule that contains the sugar ribose Nucleic acid hybridization
instead of the deoxyribose found in DNA and the pyrimidine uracil DNA sequencing
in place of thymine. Minimal residual disease
RNA polymerase recognizes a sequence of deoxyribonucleotides Molecular testing allows more sensitive assessment of therapeutic
called the promoter within DNA. efficacy and the presence of residual diseased cells.
RNA polymerase separates the DNA strands and begins adding Molecular testing permits clinicians to make more accurate thera-
ribonucleotides, forming an initial RNA transcript consisting of peutic and prognostic decisions.
introns and exons. Real-time quantitative PCR allows assessment of the level of
Proteins function as structural components of the cell, as enzymes disease remaining, not just its presence or absence.
involved in metabolism or regulation, as receptors to regulate
cellular functions, or as antibodies for the immune system. Now that you have completed this chapter, go back and
Mutation within a gene ultimately alters the protein produced, read again the case study at the beginning and respond
which often affects the function of the protein. to the questions presented.

R E V I E W Q UESTIONS
1. If the DNA nucleotide sequence is 5-ATTAGC-3, then the 5. A 40-year-old patient enters the hospital with a rare form
mRNA sequence transcribed from this template is: of cancer caused by faulty cell division regulation. This
a. 5-GCUAAU-3 cancer localized in the patients spleen. An ambitious labo-
b. 5-AUUAGC-3 ratory developed a molecular test to verify the type of
c. 5-TAATCG-3 cancer present. This molecular test would require patient
d. 5-UAAUCG-3 samples taken from which two tissues?
a. Abnormal growths found on the skin and in the bone
2. Cells with damaged DNA and mutated or nonfunctioning marrow
p53: b. Normal splenic tissue and cancerous tissue
a. Are arrested in G1 and the DNA is repaired c. Cancerous tissue in spleen and bone marrow
b. Continue to divide, which leads to tumor progression d. Peripheral blood and cancerous tissue in the spleen
c. Divide normally, producing identical daughter cells
d. Go through apoptosis 6. One main difference between PCR and RT-PCR is that:
a. PCR requires primers
3. To start DNA replication, DNA polymerase requires an b. PCR uses reverse transcriptase to elongate the primers
available 3 hydroxyl group found on the: c. RT-PCR forms billions of cDNA fragments
a. Leading strand d. RT-PCR requires ligase to amplify the target DNA
b. mRNA
c. Parent strand 7. Which one of the following statements about gel electro-
d. Primer phoresis is false?
a. The gel is oriented in the chamber with the wells at
4. Ligase joins Okazaki fragments of the: the positive terminal.
a. 5-to-3 template strand b. A buffer solution is required to maintain the electrical
b. Lagging strand current.
c. Leading strand c. The matrix of a polyacrylamide gel is tighter than that
d. Primer fragments of an agarose gel.
d. The larger DNA fragments will be closest to the wells
of the gel.
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 485

8. Autoradiography of DNA is the: 10. Which of the following statements about minimal residual
a. Detection of radioactive deoxyribonucleotides disease is true?
b. Exposure of the gel to UV light a. Clinical remission of hematologic cancers can be
c. Transfer of DNA to a nitrocellulose filter determined by molecular techniques such as PCR and
d. Use of EtBr to visualize the DNA banding pattern flow cytometry.
b. Real-time quantitative PCRdetermined copy number
9. In a test for B-cell gene rearrangement, the pattern seen in of BCR/ABL rearrangements will always be lower in
the patients sample is a smear. This smear indicates that: molecular remission than in clinical remission.
a. A monoclonal population of B cells exists c. Qualitative PCR that uses a known copy number of a
b. A polyclonal population of B cells exists target sequence is of use in determining minimal
c. Contamination of the sample occurred residual disease levels.
d. Too much probe was used during detection d. Minimal residual disease assessment can aid
physicians in making treatment decisions but does
not yet offer insights into prognosis.

REFERENCES
1. Kaufman RE: Synthesis of sickle and normal hemoglobins: 17. Marks DI, Kurz BW, Link MP, et al: Altered expression of p53
laboratory and clinical implications. Lab Med 30:129-133, and mdm-2 proteins at diagnosis is associated with early
1999. treatment failure in childhood acute lymphoblastic leuke-
2. Mayo MM, Samuel SM: Clinical pathology rounds: diagnos- mia. J Clin Oncol 15:1158-1162, 1997.
ing hemoglobin SC. Lab Med 30:98-101, 1999. 18. Gustafsson B, Stal O, Gustafsson B: Overexpression of MDM2
3. International Human Genome Sequencing Consortium: in acute childhood lymphoblastic leukemia. Pediatr Hematol
Finishing the euchromatic sequence of the human genome. Oncol 15:519-526, 1998.
Nature 431:931-945, 2004. 19. Zhou M, Gu L, Abshire TC, et al: Incidence and prognostic
4. Calladine CR, Drew HR, Luisi BF: Understanding DNA: the significance of MDM2 oncoprotein overexpression in relapsed
molecule and how it works, ed 3, San Diego, 2004, Academic childhood acute lymphoblastic leukemia. Leukemia 14:61-67,
Press, pp 1-17. 2000.
5. Mattick JS, Makunin IV: Non-coding RNA. Hum Mol Genet 20. Bartlett JMS, White A: Extraction of DNA from whole blood.
15:R17-R29, 2006. In Bartlett JMS, Stirling D, editors: PCR protocols, ed 2, Totowa,
6. Lewin B: Messenger RNA. In Genes IX, ed 9, Sudbury, 2008, NJ, 2003, Humana Press, pp 29-31.
Mass, Jones & Bartlett, pp 127-150. 21. Shimizu H, Burns JC: Extraction nucleic acids: sample prepa-
7. Basic molecular genetic mechanisms. In Lodish H, Berk A, ration from paraffin-embedded tissues. In Innis MA, Gelfand
Kaiser CA, et al, editors: Molecular cell biology, ed 6, New York, DH, Sninsky JJ, editors: PCR strategies, San Diego, 1995,
2008, WH Freeman, pp 111-164. Academic Press, pp 32-38.
8. Kastan MB, Onyekwere O, Sidransky D, et al: Participation of 22. Harder J, Schroder JM: RNase 7, a novel innate immune
p53 protein in the cellular response to DNA damage. Cancer defense antimicrobial protein of healthy human skin. J Biol
Res 51(23 pt 1):6304-6311, 1991. Chem 277:46779-46784, 2002.
9. Fritsche M, Haessler C, Brandner G: Induction of nuclear 23. Tang YW, Procop GW, Persing DH: Molecular diagnostics of
accumulation of the tumor-suppressor protein p53 by DNA- infectious diseases. Clin Chem 11:2021-2038, 1997.
damaging agents. Oncogene 8:307-318, 1993. 24. Miller WH, Kakizuka A, Frankel SR, et al: Reverse transcrip-
10. Lane DP: p53, guardian of the genome. Nature 358:15-16, tion polymerase chain reaction for the rearranged retinoic
1992. acid receptor clarifies diagnosis and detects minimal resid-
11. Kelman Z, Prokocimer M, Peller S, et al: Rearrangements in ual disease in acute promyelocytic leukemia. Proc Natl Acad
the p53 gene in Philadelphia chromosome positive chronic Sci U S A 89:2694-2698, 1992.
myelogenous leukemia. Blood 74:2318-2324, 1989. 25. Schwartz RC, Sonenshein GE, Bothwell A, et al: Multiple
12. Rovira A, Urbano-Ispizua A, Cervantes F, et al: p53 tumor expression of Ig -chain encoding RNA species in murine
suppressor gene in chronic myelogenous leukemia: a sequen- plasmacytoma cells. J Immunol 126:2104-2108, 1981.
tial study. Ann Hematol 70:129-133, 1995. 26. Mullis KB, Faloona FA: Specific synthesis of DNA in vitro
13. Beck Z, Kiss A, Toth FD, et al: Alterations of P53 and RB genes via a polymerase-catalyzed chain reaction. Methods Enzymol
and the evolution of the accelerated phase of chronic myeloid 155:335-350, 1987.
leukemia. Leuk Lymphoma 38:587-597, 2000. 27. Saiki RK, Scharf S, Faloona F, et al: Enzymatic amplification
14. Cano I, Martinez J, Quevedo E, et al: Trisomy 12 and p53 of -globin genomic sequences and restriction site analysis
deletion in chronic lymphocytic leukemia detected by fluo- for diagnosis of sickle cell anemia. Science 230:1350-1354,
rescence in situ hybridization: association with morphology 1985.
and resistance to conventional chemotherapy. Cancer Genet 28. Studwell PS, ODonnell M: Processive replication is contin-
Cytogenet 90:118-124, 1996. gent on the exonuclease subunit of DNA polymerase III holo-
15. Amiel A, Arbov L, Manor Y, et al: Monoallelic p53 deletion enzyme. J Biol Chem 265:1171-1178, 1990.
in chronic lymphocytic leukemia detected by interphase cyto- 29. Saiki RK, Gelfand DH, Stoffel S, et al: Primer-directed enzy-
genetics. Cancer Genet Cytogenet 97:97-100, 1997. matic amplification of DNA with a thermostable DNA poly-
16. Lazaridou A, Miraxtsi C, Korantzis J, et al: Simultaneous merase. Science 239:487-491, 1988.
detection of BCL-2 protein, trisomy 12, retinoblastoma and 30. Wiedbrauk DL: Molecular methods for virus detection. Lab
P53 monoallelic gene deletions in B-cell chronic lymphocytic Med 23:737-742, 1992.
leukemia by fluorescence in situ hybridization (FISH): rela- 31. Altschul SF, Gish W, Miller W, et al: Basic local alignment
tion to disease status. Leuk Lymphoma 36:503-512, 2000. search tool. J Mol Biol 215:403-410, 1990.
486 PART V Leukocyte Disorders

32. Mifflin TE: Setting up a PCR laboratory. In Dieffenbach CW, 51. Yuen E, Brown RD: Southern blotting of IgH rearrangements
Dveksler GS, editors: PCR primer: a laboratory manual, ed 2, in B-cell disorders. In Brown RD, Ho P, editors: Multiple
Cold Spring Harbor, NY, 2003, Cold Spring Harbor Labora- myeloma, Totowa, NJ, 2005, Humana Press, pp 85-103.
tory Press, pp 5-14. 52. Dowton SB, Slaugh RA: Diagnosis of human heritable dis-
33. Najfeld V: Conventional and molecular cytogenic basis of eases: laboratory approaches and outcomes. Clin Chem
hematologic malignancies. In Hoffman R, Benz EJ Jr, Shattil 41:785-794, 1995.
SJ, et al, editors: Hematology: basic principles and practice, ed 5, 53. Cossman J, Uppenkamp M, Sundeen J, et al: Molecular genet-
Philadelphia, 2009, Churchill Livingstone, pp 791-838. ics and the diagnosis of lymphoma. Arch Pathol Lab Med
34. McClure JS, Litz CE: Chronic myelogenous leukemia: molecu- 112:117-127, 1988.
lar diagnostic considerations. Hum Pathol 25:594-597, 1994. 54. Gill JI, Gulley ML: Immunoglobulin and T-cell receptor gene
35. Keung YK, Beaty M, Powell BL, et al: Philadelphia chromo- rearrangement. Diagn Hematol 8:751-770, 1994.
some positive myelodysplastic syndrome and acute myeloid 55. Griesser H, Tkachuk D, Reis D, et al: Gene rearrangements
leukemiaretrospective study and review of literature. Leuk and translocations in lymphoproliferative diseases. Blood
Res 28:579-586, 2004. 73:1402-1415, 1989.
36. Bartram CR, de Klein A, Hagemeijer A, et al: Translocation 56. Noorali S, Pervez S, Moatter T, et al: Characterization of
of the c-abl oncogene correlates with the presence of a T-cell non-Hodgkins lymphoma and its association with
Philadelphia chromosome in chronic myelocytic leukemia. Epstein-Barr virus in Pakistani patients. Leuk Lymphoma
Nature 306:277-280, 1983. 44:807-813, 2003.
37. Heisterkamp N, Stephenson JR, Groffen J, et al: Localization 57. Kurosu K, Yumoto N, Mikata A, et al: Monoclonality of B-cell
of the c-abl oncogene adjacent to a translocation break point lineage in primary pulmonary lymphoma demonstrated by
in chronic myelocytic leukemia. Nature 306:239-242, 1983. immunoglobulin heavy chain gene sequence analysis of his-
38. Clark SS, McLaughlin J, Crist WM, et al: Unique forms of the tologically non-definitive transbronchial biopsy specimens.
abl tyrosine kinase distinguish Ph1-positive CML from Ph1- J Pathol 178:316-322, 1996.
positive ALL. Science 235:85-88, 1987. 58. Hanson CA: Clinical applications of molecular biology in
39. Preudhomme C, Revillion F, Merlat A, et al: Detection of diagnostic hematopathology. Lab Med 24:562-573, 1993.
BCR-ABL transcripts in chronic myeloid leukemia (CML) 59. Goto M, Oka S, Okuzumi K, et al: Evaluation of acridinium-
using a real time quantitative RT-PCR assay. Leukemia esterlabeled DNA probes for identification of Mycobacterium
13:957-964, 1999. tuberculosis and Mycobacterium aviumMycobacterium intracel-
40. Hebert J, Cayuela JM, Daniel MT, et al: Detection of minimal lulare complex in culture. J Clin Microbiol 2473-2476, 1991.
residual disease in acute myelomonocytic leukemia with 60. Leary JJ, Brigati DJ, Ward DC: Rapid and sensitive colorimet-
abnormal marrow eosinophils by nested polymerase chain ric method for visualizing biotin-labeled DNA probes hybrid-
reaction with allele specific amplification. Blood 84:2291- ized to DNA or RNA immobilized on nitrocellulose: Bio-blots.
2296, 1994. Proc Natl Acad Sci U S A 80:4045-4049, 1983.
41. Sambrook J, Russell DW: Gel electrophoresis of DNA and 61. Morrell JI, Greenberger LM, Pfaff DW: Comparison of horse-
pulsed-field agarose gel electrophoresis. In Sambrook J, radish peroxidase visualization methods: quantitative results
Russell DW, editors: Molecular cloning: a laboratory manual, and further technical specifics. J Histochem Cytochem 29:903-
ed 3, Cold Spring Harbor, NY, 2001, Cold Spring Harbor 916, 1981.
Laboratory Press, pp 5.4-5.86. 62. Thorpe GH, Kricka LJ: Enhanced chemiluminescent reactions
42. Protein structure and function. In Lodish H, Berk A, Kaiser catalyzed by horseradish peroxidase. Methods Enzymol 133:
CA, et al, editors: Molecular cell biology, ed 6, New York, 2007, 331-353, 1986.
WH Freeman, pp 63-110. 63. Wilding P: Miniaturization: DNA chips and devices. In Bruns
43. Nathans D, Smith HO: Restriction endonucleases in the anal- DE, Ashwood ER, Burtis CA, editors: Fundamentals of molecu-
ysis and restructuring of DNA molecules. Annu Rev Biochem lar diagnostics, St Louis, 2007, Saunders.
44:273-293, 1975. 64. Buckingham L. DNA sequencing. In Buckingham L, Flaws
44. Ross DW: Restriction enzymes. Arch Pathol Lab Med 114:906, ML, editors: Molecular diagnostics: fundamentals, methods, and
1990. clinical applications, Philadelphia, 2007, FA Davis.
45. De Stefano V, Finazzi G, Mannucci PM: Inherited thrombo- 65. Sanger F, Nicklen S, Coulson AR: DNA sequencing with
philia: pathogenesis, clinical syndromes, and management. chain-terminating inhibitors. Proc Natl Acad Sci U S A
Blood 87:3531-3544, 1996. 74:5463-5467, 1977.
46. Liu XY, Nelson D, Grant C, et al: Molecular detection of a 66. van der Velden VHJ, Hochhaus A, Cazzaniga G, et al: Detec-
common mutation in coagulation factor V causing thrombo- tion of minimal residual disease in hematologic malignan-
sis via hereditary resistance to activated protein C. Diagn Mol cies by real-time quantitative PCR: principles, approaches,
Pathol 4:191-197, 1995. and laboratory aspects. Leukemia 17:1013-1034, 2003.
47. Southern EM: Detection of specific sequences among DNA 67. Schlesinger J, Tnjes M, Schueler M, et al: Evaluation of the
fragments separated by gel electrophoresis. J Mol Biol 98:503- LightCycler 1536 instrument for high-throughput quantita-
517, 1975. tive real-time PCR. Methods 50:S19-S22, 2010.
48. Chen CY, Shiesh SC, Wu SJ: Rapid detection of K-ras muta- 68. Le Guillou-Guillemette H, Lunel-Fabiani F: Detection and
tions in bile by peptide nucleic acid-mediated PCR clamping quantification of serum or plasma HCV RNA: mini review of
and melting curve analysis: comparison with restriction frag- commercially available assays. Methods Mol Biol 510:3-14,
ment length polymorphism analysis. Clin Chem 50:481-489, 2009.
2004. 69. Ahmad AI, Ghasemi JB: New FRET primers for quantitative
49. Saiki RK, Walsh PS, Levenson CH, et al: Genetic analysis of real-time PCR. Anal Bioanal Chem 387:2737-2743, 2007.
amplified DNA with immobilized sequence-specific oligo- 70. Luthra R, Medeiros LJ: TaqMan reverse transcriptase-
nucleotide probes. Proc Natl Acad Sci U S A 86:6230-6234, polymerase chain reaction coupled with capillary electropho-
1989. resis for quantification and identification of bcr-abl transcript
50. Thomas PS: Hybridization of denatured RNA and small DNA type. Methods Mol Biol 335:135-145, 2006.
fragments transferred to nitrocellulose. Proc Natl Acad Sci 71. Kern W, Voskova D, Schoch C, et al: Determination of relapse
U S A 77:5201-5205, 1980. risk based on assessment of minimal residual disease during
CHAPTER 32 Molecular Diagnostics in the Clinical Laboratory 487
complete remission by multiparameter flow cytometry in 76. Tan W, Wang K, Drake TJ: Molecular beacons. Curr Opin Chem
unselected patients with acute myeloid leukemia. Blood Biol 8:547-553, 2004.
104:3078-3085, 2004. 77. Stentoft J, Pallisgaard N, Kjeldsen E, et al: Kinetics of BCR-
72. Fenk R, Ak M, Kobbe G, et al: Levels of minimal residual ABL fusion transcript levels in chronic myeloid leukemia
disease detected by quantitative molecular monitoring herald patients treated with ST1571 measured by quantitative real-
relapse in patients with multiple myeloma. Haematologica time polymerase chain reaction. Eur J Haematol 67:302-308,
89:557-566, 2004. 2001.
73. Osborne D, Frost L, Tobal K, et al: Elevated levels of WT1 78. Gabert J, Beillard E, van der Velden VH, et al: Standardization
transcripts in bone marrow harvest are associated with a high and quality control studies of real-time quantitative reverse
relapse risk in patients autografted for acute myeloid leuke- transcriptase polymerase chain reaction of fusion transcripts
mia. Bone Marrow Transplant 36:67-70, 2005. for residual disease detection in leukemiaa Europe Against
74. Brggemann M, Schrauder A, Raff T, et al: European Working Cancer program. Leukemia 17:2318-2357, 2003.
Group for Adult Acute Lymphoblastic Leukemia (EWALL); 79. Dadi S, Le Noir S, Asnafi V, et al: Normal and pathological
International Berlin-Frankfurt-Mnster Study Group (I-BFM- V(D)J recombination: contribution to the understanding of
SG): Standardized MRD quantification in European ALL human lymphoid malignancies. Adv Exp Med Biol 650:180-
trials: proceedings of the Second International Symposium 194, 2009.
on MRD assessment in Kiel, Germany, 18-20 September 80. Wittek M, Strmer M, Doerr HW, et al: Molecular assays for
2008. Leukemia 24:521-535, 2010. monitoring HIV infection and antiretroviral therapy. Expert
75. Lion T, Henn T, Gaiger A, et al: Early detection of relapse after Rev Mol Diagn 7:237-746, 2007.
bone marrow transplantation in patients with chronic
myelogenous leukaemia. Lancet 341:275-276, 1993.
33 Flow Cytometric Analysis
in Hematologic Disorders
Magdalena Czader

OUTLINE OBJECTIVES
Specimen Processing After completion of this chapter, the reader will be able to:
Flow Cytometry: Principle
and Instrumentation 1. Describe the technique of flow cytometry, including 4. Recognize the key immunophenotypic features of
Pattern Recognition specimen selection and preparation, instrumentation, normal bone marrow, peripheral blood, and lymph
Approach to Analysis of data collection, and antibody panel design. node tissue, and specimens from patients with acute
Flow Cytometric Data 2. Discuss the pattern recognition approach to analysis leukemia or lymphoma.
Concept of Gating of flow cytometric data for diagnosis and follow-up of 5. Discuss novel applications of flow cytometry beyond
Analysis of Flow hematologic malignancies. the immunophenotyping of hematologic malignancies.
Cytometric Data 3. Identify basic cell populations defined by flow cyto-
Cell Populations Identified metric parameters.
by Flow Cytometry
Granulocytic Lineage
Monocytic Lineage
Erythroid Lineage
Megakaryocytic Lineage
Lymphoid Lineage
Flow Cytometric Analysis
of Myeloid Disorders CASE STUDIES
(Acute Myeloid After studying the material in this chapter, the reader should be able to respond to the following
Leukemias and Chronic case studies:
Myeloid Disorders)
Acute Myeloid Leukemias Case 1
with Recurrent A 58-year-old man had a 5-month history of extensive right cervical lymphadenopathy and night
Cytogenetic Abnormalities sweats. His complete blood count (CBC) results were within normal limits. Physical examination
Acute Myeloid Leukemias showed additional bilateral axillary lymphadenopathy. The cervical lymph node was excised. His-
Not Otherwise Categorized tologic examination revealed nodular architecture with predominantly medium-sized lymphoid
Myeloproliferative cells with irregular nuclear outlines. Flow cytometric data are presented in Figure 33-1.
Neoplasms and
1. What cell subpopulation predominates on the forward scatter (FS)/side scatter (SS) scattergram?
Myelodysplastic
2. List antigens positive in this population.
Syndromes
Flow Cytometric Analysis 3. Does the pattern of light chain expression support the diagnosis of lymphoma?
of Lymphoid Neoplasms
(Lymphoblastic Leukemia/ Case 2
Lymphoma and Mature A 3-year-old girl was brought to the physician because of fatigue and fevers. The CBC revealed a
Lymphoid Neoplasms) WBC count of 3 109/L, Hb level of 8.3g/dL, and platelet count of 32 109/L. Review of the
B Lymphoblastic peripheral blood film showed rare undifferentiated blasts with occasional cytoplasmic blebs. No
Leukemia/Lymphoma granules or Auer rods were identified. Bone marrow examination showed a marked increase in
T Lymphoblastic blasts (79%) and decreased background trilineage hematopoiesis. Flow cytometric analysis was
Leukemia/Lymphoma performed. In addition to the markers shown in Figure 33-2, the population of interest was positive
Mature Lymphoid
for CD34, CD33, CD41, and HLA-DR.
Neoplasms
1. What abnormal features are observed on the CD45/SS scattergram?
Other Applications of
Flow Cytometry Beyond 2. What is the most likely diagnosis considering the constellation of markers expressed by the
Immunophenotyping of predominant population?
Hematologic Malignancies Continued

488
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 489

CASE STUDIEScontd
After studying the material in this chapter, the reader should be able to respond to the following case studies:

103
1023

102

CD10-PC5
101
SS

100
0

0 1023 100 101 102 103

FS CD19-FITC
A B

103 103

102 102
LAMBDA-PE
CD5-FITC

101 101

100 100

100 101 102 103 100 101 102 103

CD19-RD KAPPA-FITC
C D
Figure 33-1 Scattergrams showing immunophenotypic features of lymphoid cells from the patient in Case 1. FS, Forward scatter; SS, side scatter;
RD, rhodamine; FITC, fluorescein isothiocyanate.
Continued
490 PART V Leukocyte Disorders

CASE STUDIEScontd
After studying the material in this chapter, the reader should be able to respond to the following case studies:

1023
SS

100 101 102 103

A CD45-ECD

103

102
CD6-PC5

101

100

100 101 102 103

B CD61-PE
Figure 33-2 Predominant bone marrow population for the patient in Case 2. SS, Side scatter, ECD, phycoerythrin-Texas Red; PE, phycoerythrin.

F
low cytometry was originally designed to measure physi- over other techniques is its ability to analyze rapidly multiple
cal properties of cells based on their ability to deflect parameters in a large number of cells. When one adds the
light. Over the years, it has evolved to include detection capability of identifying and quantifying rare-event cells in a
of fluorescent signals emitted by dyes bound directly to specific heterogeneous cell population, the value of flow cytometry to
molecules or attached to proteins through monoclonal anti- clinical hematology becomes obvious. Currently, this tech-
bodies. The development of monoclonal antibodies is the most nique not only is applied to the analysis of cell lineage in acute
significant factor contributing to todays broad application of leukemia or the detection of clonality in lymphoid populations
flow cytometry. Although the term flow cytometry implies the but also makes it possible to discern abnormal populations
measurement of a cell, this technique is applied successfully in chronic myeloid neoplasms, quantitate minimal residual
to the study of other particles, including chromosomes, micro- disease, and monitor immunodeficiency states. Immunophe-
organisms, and proteins. The main advantage of flow cytometry notypes that originally were used to supplement morphologic
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 491

classification frequently correlate with specific cytogenetic or TABLE 33-1 Lineage-Associated Markers Commonly
molecular abnormalities. According to a classification of Analyzed in Routine Flow Cytometry
hematopoietic neoplasms recommended by the World Health
Lineage Markers
Organization,1 one no longer can rely solely on morphology
for diagnosis of hematologic malignancies. The optimal diag- Immature CD34
nostic algorithm integrates morphologic, immunophenotypic, CD117
and genotypic information. This approach emphasizes the
Terminal deoxynucleotidyl transferase
central role that flow cytometry plays in the hematopathology
Granulocytic/monocytic CD33
laboratory.
CD13
The discussion in this chapter focuses on the use of flow
CD15
cytometry in the routine hematopathology laboratory. The
CD14
chapter follows the life of the flow cytometric specimen that
Erythroid CD71
starts with specimen processing and ends with the final diag-
Glycophorin A
nosis. The discussion is divided into preanalytical (specimen
Megakaryocytic CD41
processing), analytical (flow cytometric instrumentation and
CD42
analysis), and postanalytical (immunophenotypic features of
CD61
hematopoietic disorders) sections.
B lymphocytes CD19
CD20
SPECIMEN PROCESSING CD22
Light chain
Flow cytometric analysis is particularly useful in diagnosing
Light chain
hematologic malignancies. The specimens most commonly
T lymphocytes CD2
analyzed are bone marrow, peripheral blood, and lymphoid
CD3
tissues. In addition, immunophenotyping is often performed
CD4
on body cavity fluids and solid tissues when they are suspected
CD5
to harbor a hematologic malignancy.2
CD7
Prolonged transport or transport under inappropriate con-
CD8
ditions may render a specimen unsuitable for analysis. Periph-
eral blood and bone marrow specimens should be processed
within 24 to 48 hours from the time of collection. Certain
specimens, such as body cavity fluids or samples from neo- antibodies to pass through the cell membrane. A predeter-
plasms with a high proliferative activity, may require even mined panel of antibodies may be used to detect membrane-
more rapid processing. bound and intracellular markers. Simultaneous analysis of
When cells are suspended in a fluid, as in peripheral blood multiple markers, known as multicolor or multiparameter flow
and bone marrow, minimal sample preparation is required. cytometry, has numerous advantages. It facilitates visualization
These specimens are collected into a tube or container with of antigen expression and maturation patterns, which are often
an anticoagulant, heparin or ethylenediaminetetraacetic acid disturbed in hematopoietic malignancies. In addition, regard-
(EDTA), and are transported to the flow cytometry laboratory less of the complexity of the specimen, analysis can be accom-
at room temperature. Bone marrow biopsy specimens and plished using a few tubes and with a lower total number of cells,
solid tissue specimens, including core biopsy samples, are sub- which saves reagents, time, and data storage. There is no con-
mitted in culture media to maintain viability or on saline- sensus on the standardized panel of antibodies to be used in
moistened gauze. Tissue fragments are mechanically dissociated routine flow cytometric evaluation. The U.S.-Canadian Consen-
to yield a cell suspension, usually by mincing with a scalpel. sus Project in Leukemia/Lymphoma Immunophenotyping rec-
To obtain a pure population of nucleated cells, red blood ommends the comprehensive approach with multiple markers
cells (RBCs) are lysed before staining. The analytical process for myeloid and lymphoid lineage.3 Selected markers com-
depends on the cellularity and viability of the specimen; both monly analyzed by flow cytometry are presented in Table 33-1.
are routinely assessed before a sample is stained. Cell count
can be obtained using automated cell counters or flow cytom-
FLOW CYTOMETRY:
etry. Trypan blue exclusion, a manual staining method, or flow
PRINCIPLE AND INSTRUMENTATION
cytometry of a specimen stained with propidium iodide or
7-amino actinomycin, is used to test viability. A cytocentrifuge The most significant discovery that led to the advancement of
slide (see Chapter 17) is prepared for the morphologic inspec- flow cytometry and its subsequent widespread application in
tion of the cell suspension. clinical practice was the development of monoclonal anti
As soon as these steps are completed, the sample is stained bodies.4 In the original hybridoma experiments, lymphocytes
with a cocktail of fluorochrome-conjugated monoclonal anti- with predetermined antibody specificity were co-cultured with
bodies. The analysis of intracytoplasmic markers requires a myeloma cell line to form immortalized hybrid cells produc-
an additional fixation and permeabilization step to allow ing specific monoclonal antibodies. For this discovery, which
492 PART V Leukocyte Disorders

TABLE 33-2 Hematolymphoid Antigens Commonly Used in Clinical Flow Cytometry


Cluster of Differentiation Function Cellular Expression
CD1a T-cell development Precursor T cells
CD2 T-cell activation Precursor and mature T cells, NK cells
CD3 Antigen recognition Precursor and mature T cells
CD4 Co-receptor for HLA class II Precursor T cells, helper T cells, monocytes
CD5 T-cell signaling Precursor and mature T cells, subset of B cells
CD7 T-cell activation Precursor and mature T cells, NK cells
CD8 Co-receptor for HLA class I Precursor T cells, suppressor/cytotoxic T cells, subset of NK cells
CD10 B-cell regulation Precursor B cells, germinal center B cells, granulocytes
CD11b Cell adhesion Granulocytic and monocytic lineage, NK cells
CD13 Unknown Granulocytic and monocytic lineage
CD14 Monocyte activation Mature monocytes
CD15 Ligand for selectins Granulocytic and monocytic lineage
CD16 Low-affinity IgG Fc receptor Granulocytic and monocytic lineage, NK cells
CD18 Cell adhesion and signaling Granulocytic and monocytic lineage
CD19 B-cell activation Precursor and mature B cells
CD20 B-cell activation Precursor and mature B cells
CD22 B-cell activation and adhesion Precursor and mature B cells
CD31 Cell adhesion Megakaryocytes, platelets, leukocytes
CD33 Cell proliferation and survival Granulocytic and monocytic lineage
CD34 Cell adhesion Hematopoietic stem cells
CD36 Cell adhesion Megakaryocytes, platelets, erythroid precursors, monocytes
CD38 Cell activation and proliferation Hematopoietic cells, including activated lymphocytes and plasma cells
CD41 Cell adhesion Megakaryocytes, platelets
CD42b Receptor for von Willebrand factor Megakaryocytes, platelets
CD45 T- and B-cell receptor activation Hematopoietic cells
CD56 Cell adhesion NK cells, subset of T cells
CD61 Cell adhesion Megakaryocytes, platelets
CD62P Homing Platelets
CD63 Cell development, activation, Platelets
growth and motility
CD64 High-affinity IgG Fc receptor Granulocytic and monocytic lineage
CD71 Iron uptake High density on erythroid precursors, low to intermediate density on
other proliferating cells
CD79a B-cell receptor signal transduction Precursor and mature B cells
CD117 Stem cell factor receptor Hematopoietic stem cells, mast cells

Ig, Immunoglobulin; NK, natural killer.

not only fueled the development of flow cytometry but also Monoclonal antibodies have various applications, includ-
had innumerable research and, more recently, clinical applica- ing immunohistochemistry, immunofluorescence, and Western
tions, Khler and Milstein received a Nobel Prize in 1984. Over blot. These methods study cellular proteins in fixed tissues or
the years, numerous antibodies were produced and tested for in cellular extracts; however, they do not provide the ability to
their lineage specificity. Categorization of these antibodies and examine antigens in their native state and cannot decipher
associated antigens is accomplished through workshops on composite cell populations with a complex antigen makeup.
human leukocyte differentiation antigens that have been held In contrast, flow cytometry can define antigen expression on
regularly since 1982. These workshops provide a forum for numerous viable cells. Currently, 17 antigens can be detected
reporting new antigens and antibodies and define a cluster of simultaneously on an individual cell.6 This is accomplished
antibodies recognizing the same antigen, called cluster of dif- by the conjugation of monoclonal antibodies to a variety of
ferentiation (CD) (Table 33-2; see also Table 33-1). Consecutive fluorochromes that can be detected directly by a flow cytom-
numbers are assigned to each new reported antigen. The Ninth eter. In a flow cytometer, particles are suspended in fluid and
International Conference on Human Leukocyte Differentiation pass one by one in front of a light source. As particles are illu-
Antigens brought to over 350 the total number of antigens minated, they emit fluorescent signals registered by detectors.
characterized.5 These results are later converted to digital output and analyzed
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 493

Sheath
fluid Excited Relaxation
state

Filters
Fluorescence
detectors

FS
detector
Laser beam

Absorption Fluorescence

SS
detector
Figure 33-3 Diagram of a flow cytometer. As cells are injected into pres-
surized sheath fluid, they are positioned in the center of the stream and one
by one exposed to the laser light. Forward scatter (FS) and side scatter (SS)
are collected by separate detectors.

using flow cytometry software. The flow cytometer consists of


fluidics, a light source (laser), a detection system, and a com- Ground Electron
puter. A brief discussion of these basic components is state
presented. Figure 33-4 Jablonski diagram showing a principle of fluorescence.
To be analyzed individually, cells must pass separately, one When electrons absorb energy, they are raised to the excited state. Subse-
by one, through the illumination and detection system of the quently, on their return to ground state, the absorbed energy is emitted in a
form of fluorescence.
flow cytometer. This passage is accomplished by injecting the
cell suspension into a stream of sheath fluid. This technique,
called hydrodynamic focusing, creates a central core of individu- the side measures side scatter (SS or SSC), which reflects surface
ally aligned cells surrounded by a sheath fluid (Figure 33-3). complexity and internal structures such as granules and vacu-
The central alignment is essential for consistent illumination oles. FS, SS, and fluorescence are displayed simultaneously on
of the cells as they pass before the laser light source. the instrument screen and registered by the computer system.
The laser is composed of a tube filled with gas, most com-
monly argon or helium-neon, and a power supply. Current is
PATTERN RECOGNITION APPROACH TO
applied to the gas to raise the electrons of the gas to the excited
ANALYSIS OF FLOW CYTOMETRIC DATA
state. When electrons return to the ground state, they emit
photons of light. Through an amplification system, a strong Concept of Gating
beam of light with light waves of identical direction, polariza- Cell populations with similar physical properties of size, cyto-
tion plane, and wavelength is produced. This narrow coherent plasmic complexity, and expression of a specific antigen form
beam of light is used to illuminate individual cells, each stained data display clusters. A gate is an electronic boundary the opera-
with antibodies conjugated to specific fluorochromes. tor uses to delineate clusters. Gating is a process of selecting,
After the absorption of laser light, the electrons of fluoro- with a cursor or computer mouse, a population of interest as
chromes are raised from the ground state to a higher energy defined by one or more flow cytometric parameters.
state (Figure 33-4). The return to the original ground level is Gating can be applied at the time of data acquisition (live
accompanied by the loss of energy, emitted as light of a specific gate) or at the time of analysis. For diagnostic purposes, data
wavelength. The flow cytometer is equipped with several pho- are collected ungated; that is, all events detected by the flow
todetectors, each specific for light of a unique color (wave- cytometer are recorded. This allows comprehensive testing and
length). The fluorescence from an individual cell is partitioned retention of positive and negative internal controls. In addi-
into its different wavelengths through a series of filters (dichroic tion, unexpected abnormal populations are detected. Gating is
mirrors) and directed to the corresponding photodetector. most commonly applied after the specimen is run through the
Fluorescent signals derived from different fluorochromes flow cytometer, when the target population is already known.
attached to particular antibodies are registered separately. In contrast, live gating focuses on the acquisition of data for a
In addition to fluorescence, scatter signals are detected. The specific cell population as defined by flow cytometric param-
detector situated directly in line with the illuminating laser eters. For example, one can collect data only on CD19+ B cells
beam measures forward scatter (FS or FSC), which is propor- to facilitate detection of a small population of monoclonal
tional to particle volume or size. A photodetector located to B cells.
494 PART V Leukocyte Disorders

1023
SS
A

0
100 101 102 103
1023

CD45-ECD
Figure 33-6 Scattergram showing differential densities of pan-
hematopoietic marker CD45 on marrow leukocytes. Lymphocytes (aqua) and
monocytes (green) show highest density of CD45 antigen. Intermediate
expression of CD45 is seen in the granulocytic population (navy) and
blasts (black). Erythroid precursors (red) are CD45. SS, Side scatter; ECD,
phycoerythrin-Texas Red.
SS

resolved further on the scattergram of CD45 antigen density


and SS (Figure 33-6). This display also provides information
on the relative proportion of specific cell populations in the
flow cytometric sample. Lymphocytes show the highest density
of CD45, with approximately 10% of the cell membrane occu-
0

pied by this antigen. Granulocytic series show intermediate


0 1023
CD45 density; erythroid precursors and megakaryocytes are
B FS
negative for CD45. The CD45/SS display is particularly useful
Figure 33-5 Main cell subpopulations of normal bone marrow. A, Bone for detection of blasts, which overlap with lymphocytes, mono-
marrow is composed of a heterogeneous population of cells of different sizes cytes, or both on the FS/SS display.7,8
and variable complexity of cytoplasm (Wright-Giemsa stain, 1000). B, Dot
The FS/SS and CD45/SS displays allow the initial identifica-
plot of forward scatter (FS, cell size) versus side scatter (SS, internal complex-
tion of the target population. Further analysis focuses on
ity) reflects the heterogeneity of bone marrow subpopulations. Lymphocytes
are smallest with negligible amount of agranular cytoplasm and are located the patterns of antigen expression, including qualitative data
closest to the origins of the axes (aqua). Monocytes are slightly larger with (antigen presence or absence) and fluorescence intensity as a
occasional granules and vacuoles (green). Granulocytic series shows promi- relative measure of surface antigen density.
nent granularity (navy).
CELL POPULATIONS IDENTIFIED
BY FLOW CYTOMETRY
Analysis of Flow Cytometric Data The surface and cytoplasmic markers expressed in hematologic
As with microscopic examination, the evaluation of flow cyto- malignancies resemble those of normal hematopoietic cell dif-
metric data is based on the inspection of visual patterns. First, ferentiation. Frequently, neoplastic cells arrest at a particular
the entire display is scanned to detect the presence of abnormal stage of development and display aberrant antigenic patterns.
populations. Subsequent analysis focuses on the antigenic The diagnosis and subclassification of hematologic neoplasms
properties of abnormal cells. is based on the knowledge of normal hematopoietic matura-
Analysis begins with inspection of dot plots presenting tion pathways.
cell size, cytoplasmic complexity, and expression of pan- In the past, the differentiation of hematopoietic cells was
hematopoietic antigen CD45. As in microscopic examination defined by morphologic criteria. Over time, it became clear that
at low magnification, the operator detects specific cell popula- specific morphologic stages of development are accompanied
tions based on their physical properties (Figure 33-5). The by distinct changes in immunophenotypes. Approximate
identification of particular populations can be confirmed and morphologic-immunophenotypic correlates exist. Because
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 495
hematopoiesis is a continuous process, the transitions between the appearance of additional glycoproteins, CD42, CD62P,
various developmental phases are not discrete. and CD63.
All hematopoietic progeny are derived from pluripotent
stem cells. These cells are morphologically unrecognizable and Lymphoid Lineage
are defined by their functional and antigenic characteristics. The B and T lymphocytes are derived from lymphoid progeni-
They usually express a combination of CD34, CD117 (c-kit), tors that express CD34, terminal deoxynucleotidyl transferase
CD38, and HLA-DR.9 As hematopoietic cells mature, they (TdT), and HLA-DR. Lymphoid differentiation is characterized
lose stem cell markers and acquire lineage-specific antigens. A by a continuum of changes in the expression of surface and
brief discussion of the maturation sequence of the major intracellular antigens. The earliest B-cell markers include CD19,
hematopoietic cell lineages is presented in the following cytoplasmic CD22, and cytoplasmic CD79.12 As B-cell precur-
sections. sors mature, they acquire the CD10 antigen. The appearance
of the mature B-cell marker CD20 coincides with the decrease
Granulocytic Lineage in CD10 antigen expression. Another specific immature B-cell
The stages of granulocytic lineage development, as defined by marker is the cytoplasmic chain that eventually is transported
the expression of specific antigens, correspond closely to the to the surface and forms the B-cell receptor. At this stage, the
morphologic sequence.10 The first morphologically recogniz- immunoglobulin chains in so-called naive B cells have become
able cell committed to the granulocytic lineage is the myelo- rearranged. The normal mature B-cell population shows a mix
blast (see Chapter 12). The myeloblast is characterized by of and light chainexpressing cells. The exclusive expres-
expression of immature cell markers CD34, CD38, HLA-DR, sion of only or molecules is a marker of monoclonality,
and stem cell factor receptor CD117. Pan-myeloid markers seen frequently in mature B-cell neoplasms. The differentiation
CD13 and CD33, present on all myeloid progeny, are first of mature naive B cells, often recapitulated by B-cell malignan-
expressed at this stage. As the myeloblast matures to a pro cies, is discussed in detail in Chapter 37.
myelocyte, it loses CD34 and HLA-DR and acquires the CD15 Similar to B-cell precursors, immature T cells express CD34
antigen. Further maturation to the myelocyte stage leads to the and TdT.13 The first markers associated with T-cell lineage
expression of CD11b, temporary loss of CD13, and a gradual include CD2, CD7, and cytoplasmic CD3. CD2 and CD7 are
decrease in the density of CD33. Finally, as granulocytic cells also present in natural killer (NK) cells and, along with
near the band stage, CD16 is acquired, and the density of the CD56 molecule, are used to detect NK cellderived neo-
CD13 increases. plasms. In T cells, the expression of CD2, CD7, and cytoplas-
mic CD3 is followed by the appearance of CD1a and CD5 and
Monocytic Lineage coexpression of CD4 and CD8 antigens. Finally, the CD3
The earliest immunophenotype stage of monocytic develop- antigen appears on the cell surface, and CD4 or CD8 is lost.
ment is defined by a gradual increase in the density of CD13, The sequential transition from double-negative (CD4CD8)
CD33, and CD11b antigens. Subsequent acquisition of CD15 through double-positive (CD4+CD8+) stages generates a popu-
and CD14 marks the transition to a promonocyte and mature lation of mature helper (CD4+) and suppressor (CD8+) T cells.
monocyte. In contrast to the granulocytic series, strong expres- T-cell differentiation occurs in the thymus.
sion of CD64 and HLA-DR antigens persists throughout mono-
cytic maturation.
FLOW CYTOMETRIC ANALYSIS OF MYELOID
DISORDERS (ACUTE MYELOID LEUKEMIAS
Erythroid Lineage
AND CHRONIC MYELOID DISORDERS)
Erythroid precursors are among the few marrow cells that do
not express pan-hematopoietic marker CD45. The earliest In myeloid malignancies, flow cytometry is used for initial
marker of erythroid differentiation is the transferrin receptor, diagnosis, follow-up, and prognostication. The specific immu-
CD71. The density of this antigen increases starting in the nophenotypes are associated with select cytogenetic abnor-
pronormoblast stage and is rapidly downregulated in reticulo- malities. Because most myeloid malignancies are stem cell
cytes.11 In contrast, glycophorin A, although present on reticu- disorders, the evaluation of blast population and maturing
locytes and erythrocytes, first appears at the basophilic myeloid component is considered mandatory. Almost invari-
normoblast stage. ably, blasts are characterized by the low-density expression of
CD45 antigen. In normal bone marrow, the blast gate includes
Megakaryocytic Lineage a relatively low number of cells showing the immature myeloid
The maturation sequence of megakaryocytes is less well immunophenotype (see Figure 33-6). In acute myeloid and
defined. CD41 and CD61, referred to as glycoprotein IIb/IIIa lymphoblastic leukemias, this region becomes densely popu-
complex, appear as the first markers of megakaryocytic differen- lated by immature cells, which reflects the increased number
tiation. These antigens are present on a small subset of of blasts seen in the bone marrow (Figure 33-7). The exact
CD34+ cells believed to represent early megakaryoblasts.9 location of the immature population on the CD45/SS displays
CD31 and CD36, although not entirely specific for megakaryo- depends on the subtype of acute myeloid leukemia (AML). In
cytic lineage, also are present on megakaryoblasts. Subsequent this chapter, the immunophenotypic features of AMLs and
maturation to megakaryocytes and platelets is characterized by chronic myeloid neoplasms are discussed in the context of the
496 PART V Leukocyte Disorders

and myeloperoxidase, are expressed. Frequently, there is asyn-


chronous coexpression of CD34 and CD15. TdT is commonly
present.
AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22);CBFB-
MYH11 is characterized by the presence of immature cells with
expression of CD34, CD117, and TdT and a subpopulation of
maturing cells showing monocytic (CD14, CD11b, CD4) and
granulocytic (CD15) markers.15 The aberrant coexpression of
CD2 on the monocytic population is common.
Acute promyelocytic leukemia, AML with t(15;17)
(q22;q12);PML-RARA, shows a specific immunophenotype. In
contrast to most less-differentiated myeloid leukemias, acute
promyelocytic leukemia manifests with high SS, which reflects
the granular cytoplasm of leukemic cells (Figure 33-9). The
constellation of immunophenotypic features used to diagnose
acute promyelocytic leukemia include lack of CD34 and
HLA-DR antigens, presence of homogeneous strong CD33
along with myeloperoxidase, and variable CD13 and CD15.16
A AMLs with t(9;11)(p22;q23);MLLT3-MLL in adults most
commonly present with monocytic differentiation. The immu-
nophenotypic features are nonspecific and can be seen in any
1023

acute myelomonocytic or monocytic leukemia (negative for


CD34 and positive for CD33, CD13, CD14, CD4, CD11b,
and CD64).

Acute Myeloid Leukemias Not


Otherwise Categorized
SS

In the least-differentiated AMLsAML with minimal differen-


tiation and AML without maturationblasts are present in the
region of low-density CD45 antigen and display low SS reflect-
ing their relatively agranular cytoplasm. Even the least differ-
entiated AML with minimal differentiation is usually positive
for myeloid markers. The expression of CD13, CD33, and
CD117 is common. Primitive hematopoietic antigens such as
CD34 and HLA-DR are often seen. Myeloperoxidase is absent
0

100 101 102 103 or is expressed in only a few cells. The immunophenotypic
profile of AML with maturation is similar, but more mature
B CD45-ECD
myeloid markers such as CD15 and myeloperoxidase are often
Figure 33-7 Bone marrow specimen showing acute leukemia. Note
expressed.
uniform cytologic and flow cytometric characteristics. A, Bone marrow aspi-
Occasionally, there is aberrant coexpression of antigens.
rate from a patient with acute lymphoblastic anemia (Wright-Giemsa stain,
500). B, CD45 versus side scatter (SS) plot shows a homogeneous popula- Simultaneous expression of early and late markers of myeloid
tion of blasts with a marked decrease in normal hematopoietic elements. differentiation on the leukemic blasts is not uncommon
Compare with the heterogeneous pattern of normal bone marrow in Figure (asynchronous antigen expression). Similarly, markers specific
33-6. ECD, Phycoerythrin-Texas Red. for other lineages such as lymphoid lineages may be seen
on myeloid blasts. The most common example is CD7
antigen, which is usually present in the T/NK cell population
World Health Organization classification, which introduced (Figure 33-10).
categories defined by recurrent cytogenetic abnormalities.1 Acute myelomonocytic leukemia and acute monoblastic
These leukemias often show specific immunophenotypes and leukemia usually show higher expression of CD45, similar to
are discussed separately in the following sections. normal monocytic precursors. In addition, in acute myelo-
monocytic leukemia, a population of primitive myeloid blasts
Acute Myeloid Leukemias with Recurrent is often seen (Figure 33-11). The expression of myeloid markers
Cytogenetic Abnormalities and antigens associated with monocytic lineage, such as CD14,
In most cases, AML with t(8;21)(q22;q22);RUNX1-RUNX1T1 CD4, CD11b, and CD64, is commonly seen. Although CD14
shows an immature myeloid immunophenotype with high- is present on all mature monocytes, it may be absent in mono-
density CD34 and coexpression of CD19 (Figure 33-8).14 In cytic leukemias.17 More immature monocytic markers, such as
addition, numerous myeloid antigens, including CD33, CD13, CD64, are more consistently expressed.
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 497

103

1023

102

CD33-PE
SS

101

100
0

100 101 102 103 100 101 102 103

A CD45-ECD B CD45-ECD

103

102
CD34-PC5

101

100

100 101 102 103

CD19-FITC
C
Figure 33-8 Acute myeloid leukemia with t(8;21)(q22;q22);RUNX1-RUNX1T1. A, CD45 versus side scatter (SS) showing increase in blasts (red) with residual
lymphocytes (aqua). B and C, Blasts are positive for CD33 and CD34 with characteristic coexpression of CD19 antigen. ECD, Phycoerythrin-Texas Red; FITC,
fluorescein isothiocyanate.

Acute erythroid leukemias are categorized into two sub- cell markers CD34 and HLA-DR on the population of leukemic
types: pure erythroid leukemia and erythroleukemia (erythroid/ megakaryoblasts varies.
myeloid leukemia). In the latter, as in acute myelomonocytic
leukemia, primitive myeloid blasts and erythroid precursors Myeloproliferative Neoplasms and
are present. Erythroid markers, CD71, glycophorin A, and Myelodysplastic Syndromes
hemoglobin (Hb), are seen in leukemic cells. In more imma- The knowledge of antigen expression in the normal differentia-
ture erythroid leukemias, glycophorin A and hemoglobin may tion of myeloid lineages allows the definition of the aberrant
be absent. In these cases, the diagnosis is based on the absence expression patterns frequently seen in chronic myeloid disor-
of myeloid markers, high expression of CD71, and scatter ders. The abnormalities detected by flow cytometry reflect mor-
characteristics. phologic features of the disorder (e.g., hypogranulation of
Acute megakaryoblastic leukemia usually shows low SS and mature neutrophils in myelodysplastic syndrome detected by
low to absent CD45. Early megakaryocytic markers, CD41 and low SS) and changes in antigen expression. Qualitative changes
CD61, are frequently expressed.18 Occasionally, the late mega- (presence or absence of a particular antigen) and quantitative
karyocytic marker CD42 is present. The expression of the stem changes (differences in the number of antigen molecules) can
1023
SS

B
0

100 101 102 103


A
CD45-ECD

103 103

102 102
HLADR-PC5

CD56-PC5
101 101

100 100

100 101 102 103 100 101 102 103


C D
CD33-PE CD34-PE
Figure 33-9 Acute promyelocytic leukemia. A, Typical side scatter (SS) pattern in acute promyelocytic leukemia corresponding to prominent granularity of
leukemic cells (red). Residual lymphocytes are shown in aqua. B, Numerous leukemic promyelocytes with distinct granules and occasional Auer rods (Wright-
Giemsa stain, 1000). C, Leukemic cells show high-density expression of CD33 antigen and lack HLA-DR. D, Similarly, CD34 antigen is absent or present
in only a few leukemic cells. SS, Side scatter; ECD, phycoerythrin-Texas Red; PE, phycoerythrin; PC5, phycoerythrin-cyanine 5.

103

102
CD13-PC5

101

100

100 101 102 103

CD7-PE
Figure 33-10 Myeloblasts of acute myeloid leukemia show aberrant coexpression of CD7 antigen. PE, Phycoerythrin; PC5, phycoerythrin-cyanine 5.
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 499

103

1023
102

CD34-PE
SS

101

100
0

100 101 102 103 100 101 102 103

CD45-ECD CD38-FITC
A B
103 103

102 102
CD11b-PC5

CD64-PC5

101 101

100 100

100 101 102 103 100 101 102 103

C CD15-FITC D CD14-FITC
Figure 33-11 Peripheral blood immunophenotyping in acute myelomonocytic leukemia. A, CD45 versus side scatter (SS) display shows myeloid blasts
(red) and a monocytic population (green). B through D, Primitive leukemic blasts are positive for CD34 and negative for CD14. In contrast, monocytic popula-
tion does not express CD34 and shows positivity for mature monocyte marker CD14 and characteristic monocytic pattern of CD11b and CD15 expression.
ECD, Phycoerythrin-Texas Red; FITC, fluorescein isothiocyanate; PC5, phycoerythrin-cyanine 5.

be used for diagnostic purposes. The interested reader is referred CD15 on myeloblasts. Asynchronous coexpression of markers
to review articles discussing the details of immunopheno can be seen in monocytic and erythroid lineages. Aberrant
typing in myelodysplastic syndromes and myeloproliferative immunophenotypes are seen in 98% of cases of myelodysplas-
neoplasms.19-21 A few examples are highlighted to illustrate the tic syndrome. More importantly, immunophenotypic abnor-
role of flow cytometry in diagnosing these diseases. malities can be seen in cases with minimal or no morphologic
SS abnormalities related to hypogranulated neutrophils are dysplasia.19 Other studies underscore the significance of immu-
seen in approximately 70% of myelodysplastic syndromes nophenotypic abnormalities in predicting the outcome after
(Figure 33-12). In high-grade myelodysplastic syndrome and stem cell transplantation.22,23
myeloproliferative neoplasms undergoing transformation, the The utility of flow cytometry in myeloproliferative neo-
increase in immature cells is detected easily. Blasts have a plasms is less well established. Specifically, the application of
variety of aberrant immunophenotypic features, most com- flow cytometry as a diagnostic tool in chronic myelogenous
monly coexpression of CD7 and CD56 antigens. Blasts and leukemia is limited to the accelerated phase or blast crisis,
maturing granulocytic precursors may show asynchronous in which a lineage of an expanding blast population needs
expression of myeloid markers, including retention of CD34 to be determined. In the chronic phase, the presence of the
and HLA-DR in late stages of granulocytic lineage or late Philadelphia chromosome as seen with conventional karyo
myeloid markers presenting early in differentiation, such as typing or by fluorescence in situ hybridization remains the
500 PART V Leukocyte Disorders

lineage-associated markers corresponding with specific stages


1023 of lymphoid development. No single marker can be used for
lineage assignment, so diagnosis is typically based on the pres-
ence of several B-cell or T-cell antigens. The sentinel feature of
mature B and T cells is the presence of surface receptor com-
plexes. The immune system responds to a wide array of anti-
gens; in healthy individuals, B and T cells express a great
diversity of surface immunoglobulin and T-cell receptor com-
SS

plexes (polyclonal populations). A neoplastic lymphoid popu-


lation is characterized by the monoclonal expression of a single
B- or T-cell receptor. In most cases, clonality confirms the
malignant nature of lymphoid proliferation. In contrast, lym-
phoid precursors are generally negative for surface immuno-
globulin and T-cell receptors and instead carry immature
markers. In lymphoblastic (precursor-derived) neoplasms, the
0

expansion of a population with homogeneous marker expres-


A 100 101 102 103
sion, rather than clonality, is diagnostic of malignancy. The
CD45-ECD following section presents the key immunophenotypic features
of lymphoblastic leukemias and lymphomas. Selected exam-
ples of the association between the immunophenotype and the
genotype are discussed.

B Lymphoblastic Leukemia/Lymphoma
B lymphoblastic leukemia/lymphoma (B-LL) is also referred to
as B acute lymphoblastic leukemia or B lymphoblastic lym-
phoma. It presents with CD19, cytoplasmic or membranous
CD22, CD79a, HLA-DR, and TdT (Figure 33-13). The expres-
sion of CD34 and CD10 is frequently seen. Surface immuno-
globulin light chains are not present. Cytoplasmic chain or
surface immunoglobulin M may be detected, however. Because
B-LL can arise at any stage of B-cell differentiation, the presence
of several specific markers usually defines early precursor, inter-
mediate, and pre-B stages. Frequently, immunophenotypes
correlate with specific cytogenetic and clinical features. In
routine practice, confirmation of cytogenetic abnormality
B using conventional karyotyping or molecular techniques is
necessary.
Figure 33-12 A, Low side scatter (SS) of hypogranular neutrophils seen
in most cases of myelodysplastic syndrome (navy). B, Corresponding photo-
micrograph of markedly dysplastic, hypogranulated neutrophils in myelodys- B Lymphoblastic Leukemia/Lymphoma with
plastic syndrome (Wright-Giemsa stain, 1000). ECD, Phycoerythrin-Texas t(v;11q23);MLL Rearranged
Red. MLL gene rearrangements occur most frequently in infant B-LL.
Unlike in most B-LLs, blasts in this leukemia are negative for
CD10 antigen.24 CD19, CD34, TdT, and occasional myeloid
defining feature of the disease. Other myeloproliferative neo- markers are present. The more mature B-cell marker CD20
plasms are not well studied. In general, flow cytometric abnor- is absent.
malities are seen in most cases manifesting cytogenetic
abnormalities.20 No consistent set of abnormalities has been B Lymphoblastic Leukemia/Lymphoma with
described, however, that can be routinely used in the workup t(9;22)(q34;q11.2);BCR-ABL1
of myeloproliferative states. Philadelphia chromosome, t(9;22);BCR-ABL1, is a hallmark of
chronic myelogenous leukemia but also can occur in pediatric
and adult B-LL. These cases are associated with a particularly
FLOW CYTOMETRIC ANALYSIS OF
dismal prognosis, so it is important to identify them promptly.
LYMPHOID NEOPLASMS (LYMPHOBLASTIC
Most BCR-ABL1positive cases have a classic intermediate
LEUKEMIA/LYMPHOMA AND MATURE
or common B-LL immunophenotype with the expression of
LYMPHOID NEOPLASMS)
CD19, CD10, CD34, and TdT. The expression of myeloid
As with the diagnosis of myeloid neoplasms, the diagnosis markers CD13 and CD33 and lack or decreased expression of
of lymphoid malignancies relies on the expression of CD38 antigen on leukemic blasts are common. The density of
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 501

103
1023

102

CD34-PC5
SS

101

100
0

100 101 102 103 100 101 102 103

A CD45-ECD B CD19-FITC

103 103

102 102
LAMBDA-PE
CD10-RD1

101 101

100 100

100 101 102 103 100 101 102 103

C CD19-FITC D KAPPA-FITC
Figure 33-13 Precursor B acute lymphoblastic leukemia. A, Low-density CD45 antigen characteristic of the blast population. B, Uniform expression of
CD34 and CD19 on leukemic blasts. C, High-density CD10 on CD19+ blasts. D, Lack of surface and light chains signifies immature B-cell population.
SS, Side scatter; ECD, phycoerythrin-Texas Red; FITC, fluorescein isothiocyanate; PE, phycoerythrin; PC5, phycoerythrin-cyanine 5.

antigens and their homogeneous or heterogeneous expression CD5, CD1a, CD4, and CD8. Usually a series of these antigens
within leukemic populations correlates closely with the pres- is detected, recapitulating the T-cell differentiation (Figure
ence of BCR-ABL1.25 33-14). CD34 and CD10 also may be present. As in other
lymphoid neoplasms, the panel of markers determines the
T Lymphoblastic Leukemia/Lymphoma lineage of the case.
T lymphoblastic leukemia/lymphoma (T-LL) is derived from
immature cells committed to T-cell lineage. Designation of Mature Lymphoid Neoplasms
leukemia or lymphoma depends on the primary site of involve- B- and T-cell lymphomas display immunophenotypes resem-
ment, bone marrow or lymph node. T-LL expresses a combina- bling their normal counterparts. The immunophenotypic fea-
tion of markers reflecting the stage of T-cell differentiation. tures of lymphomas are discussed in detail in Chapter 37 and
CD3 is the most specific T-cell marker. As with normal T cells, are summarized in Table 37-2. The flow cytometric workup of
this antigen is seen initially in the cytoplasm before appearing lymphomas is facilitated by the clonal origin of mature lym-
on the cell surface. Other T-cell antigens include CD2, CD7, phoid neoplasms, which implies that the malignant lymphoma
502 PART V Leukocyte Disorders

103
1023

102

CD3-PC5
SS

101

100
0

100 101 102 103 100 101 102 103

A CD45-ECD B CD5-FITC

103 103

102 102
CYTO CD3-PE

CD4-PE

101 101

100 100

100 101 102 103 100 101 102 103

C CD1-FITC D CD8-FITC
Figure 33-14 Precursor T acute lymphoblastic leukemia. A, Predominant population in the blast gate. B and C, Although CD3 antigen is absent from the
surface of leukemic cells, it is present in blast cytoplasm, confirming the precursor T-cell origin of the leukemia (C). Note residual normal T cells (aqua) positive
for surface CD3 and CD5 antigens. D, Simultaneous expression of CD4 and CD8 antigens. SS, Side scatter; ECD, phycoerythrin-Texas Red; FITC, fluorescein
isothiocyanate; PE, phycoerythrin; PC5, phycoerythrin-cyanine 5.

population is derived from a single cell. Therefore, all neoplas- exclusively or , is seen in most B-cell lymphomas (Figure
tic cells typically show similar genetic and immunophenotypic 33-15, B). Light chain monoclonality along with the expression
features. This stands in strong contrast to the variable immuno- of panB-cell markers is diagnostic of B-cell lymphoma. Rarely,
phenotypes of a normal lymphoid population, reflecting a lymphomas may lose the expression of surface light chains, a
process of antigen-driven selection. feature not seen in normal mature B cells.26 In most cases of
plasma cell myeloma, neoplastic plasma cells lack surface immu-
Mature B-Cell Neoplasms noglobulin light chains and express only cytoplasmic or .
Normal precursor B cells randomly rearrange immunoglobulin
heavy and light chain genes. As a result, a mature B-cell popula- Mature T-Cell Neoplasms
tion expresses a mix of heavy and light chains (Figure 33-15, In T cells, as in B cells, clonality in most cases indicates malig-
A). In contrast, a monoclonal surface light chain expression, nancy. In the past, the clonality of T cells could be only
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 503

103 10 3

102 10 2
LAMBDA-PE

LAMBDA-PE
101 10 1

100 10 0

100 101 102 103 10 0 10 1 10 2 10 3

A KAPPA-FITC B KAPPA-FITC
Figure 33-15 Comparison of surface light chain expression in reactive and malignant B cells. A, Reactive B cells show heterogeneous expression of
and . B, B-cell lymphomas are monoclonal, with the entire lymphoma population expressing only one type of light chain. FITC, Fluorescein isothiocyanate;
PE, phycoerythrin.

103 103

102 102
CD8-PC5

CD7-PE

101 101

100 100

100 101 102 103 100 101 102 103

A CD4-FITC B CD4-FITC
Figure 33-16 Mycosis fungoides. A, T-cell population is positive for CD4 antigen (red). B, Neoplastic T cells show loss of CD7. Red represents CD4 and
the expression of CD7 (green) is very low. FITC, Fluorescein isothiocyanate; PE, phycoerythrin; PC5, phycoerythrin-cyanine 5.

confirmed using the molecular analysis of T-cell receptor genes. is characterized by a mature T-cell immunophenotype with
Recently a flow cytometric assay has been shown to detect expression of CD2, surface CD3, CD5, and CD4, and a loss of
clonality in most cases of T-cell lymphoma.27 This technique the CD7 antigen (Figure 33-16). Over the years it has been
uses a broad array of antibodies against variable regions of shown that the aberrant immunophenotype is a reliable diag-
T-cell receptors. Because this methodology is not widely avail- nostic feature when the neoplastic population is sizeable.
able, often the diagnosis of T-cell lymphoma is based on aber- However, small numbers of T cells with unusual antigen
rant immunophenotype. In most cases, a loss or atypical makeup appear in inflammatory conditions28; thus the aber-
expression of a lymphoid marker can be shown using flow rant immunophenotype alone cannot be considered pathog-
cytometry. For example, mycosis fungoides/Szary syndrome nomonic of T-cell malignancy.
504 PART V Leukocyte Disorders

immunodeficiency states, diagnosis of paroxysmal nocturnal


OTHER APPLICATIONS OF
hemoglobinuria (PNH), stem cell enumeration, cell cycle anal-
FLOW CYTOMETRY BEYOND
ysis, detection of fetal hemoglobin, and monitoring of sepsis.
IMMUNOPHENOTYPING OF
Primary (inherited) and secondary (acquired) immuno
HEMATOLOGIC MALIGNANCIES
deficiency may be diagnosed using flow cytometry. The loss of
The immunophenotyping of hematolymphoid neoplasms is specific antigens (e.g., CD11/CD18 in leukocyte adhesion defi-
one of many applications of flow cytometry. Other common ciency) and functional defects (e.g., oxidative burst evaluation
applications include the diagnosis and monitoring of in chronic granulomatous disease) can be assayed.

100 101 102 103 100 101 102 103

A CD59-PE B CD59-PE

100 101 102 103

C CD59-PE
Figure 33-17 Diagnosis of paroxysmal nocturnal hemoglobinuria (PNH) is based on the decreased expression of glycosylphosphatidylinositol (GPI)linked
molecules. Different levels of GPI-anchored proteins are best visualized in red blood cells (RBCs) using an antibody against CD59 antigen. A, RBCs from a
healthy volunteer show a high number of CD59 molecules and correspond with type I cells. B, Varying percentages of type I cells (normal level of CD59
antigen) and a population with slight decrease in CD59 expression (type II cells) can be seen in PNH patients. C, Granulocytes with a complete loss of CD59
(type III cells) in a patient with PNH. This patient received numerous red blood cell transfusions; the loss of CD59 is best shown in granulocytic and monocytic
populations. PE, Phycoerythrin.
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 505

Human immunodeficiency virus infection causes a progres- Another important application of flow cytometry is cell
sive decrease in the number of CD4+ helper T cells. The abso- sorting. During sorting, a heterogeneous cell population is
lute number of helper T cells in peripheral blood correlates physically divided into subsets according to their physical or
with the stage of the disease and patient prognosis. The enu- immunophenotypic properties. High-speed sorting is achieved
meration of T cells and their subsets is easily accomplished by by charging droplets containing individual cells of interest. As
flow cytometry using antibodies against CD4 and CD8 anti- the charged droplet passes through the electrostatic field, it is
gens. The absolute numbers are derived by performing a isolated from the remainder of the sample and collected into
routine white blood cell (WBC) count on the concurrent a separate container. The primary clinical application of cell
peripheral blood specimen or by running calibrating beads sorting is in stem cell transplantation.
simultaneously with the patient sample. For years, flow cytometry remained confined to the hema-
The diagnostic approach to PNH is a prime example of how topathology and research laboratories. Currently, this method-
the application of flow cytometry increases understanding of ology is used in bone marrow transplantation, transfusion
hematologic disorders and directly contributes to clinical deci- medicine, coagulation, microbiology, molecular pathology,
sion making (see Chapter 23).29 Before the development of the and drug development. Specific examples of novel applications
flow cytometric assay, PNH diagnosis was based on detection include tissue typing, molecular testing for neoplasia-
of increased susceptibility of RBCs to lysis by the Ham or associated translocations, and follow-up of drug response,
sucrose hemolysis tests, both of which showed inconsistent such as by monitoring platelet activation after antiplatelet
sensitivity. Flow cytometry significantly improved the sensitiv- therapy.
ity and specificity of PNH testing. The absence or decreased Flow cytometry is well suited for the rapid analysis of
expression of glycosylphosphatidylinositol-anchored proteins numerous parameters. The current focus on high-throughput
on RBCs, granulocytes, and monocytes as measured by flow testing for simultaneous analysis of multiple biologic con
cytometry is diagnostic of PNH. In addition, the levels of CD59 stituents opens new avenues to the mature field of flow
expression correlate with clinical symptoms (Figure 33-17). cytometry.30

SUMMARY
Flow cytometry measures physical, antigenic, and functional prop- The immunophenotyping of hematologic specimens is based on
erties of particles suspended in a fluid. knowledge of the maturation patterns of hematopoietic cells. In
Multiparameter flow cytometry is a technique routinely used for comparison, myeloid and lymphoid malignant cells and cell popu-
the diagnosis and follow-up of hematologic disorders. lations in nonneoplastic hematologic disorders show significant
The characterization of complex specimens is achieved qualitative and quantitative differences in antigen expression.
through the analysis of individual cells for multiple parameters Flow cytometric analysis of acute leukemia determines the lineage
and the simultaneous display of data for thousands of cells. of leukemic cells. In select entities, immunophenotype corre-
The cell size, cytoplasmic complexity, and immunopheno- sponds with the underlying genetic lesion.
typic features detected by monoclonal antibodies directly con- Immunophenotyping of myelodysplastic syndromes and chronic
jugated to various fluorochromes are analyzed in clinical myeloproliferative neoplasms is an emerging application of clinical
specimens. flow cytometry.
A key starting point in flow cytometric analysis is a high-quality The clonality of mature B-cell and T-cell neoplasms can be
fresh specimen. detected by flow cytometry.
A flow cytometer consists of fluidics, a light source (laser), multiple Flow cytometric analysis is used for diagnosis and monitoring of
detectors, and a computer. immunodeficiencies, stem cell enumeration, detection of fetal
As with microscopic examination, proper evaluation of flow cyto- hemoglobin, tissue typing, molecular analysis, and drug testing.
metric data is based on the inspection of visual patterns. Initially,
the entire sample is scanned for the presence of abnormal popula- Now that you have completed this chapter, go back and
tions. Subsequently, detailed immunophenotypic features of cell read again the case studies at the beginning and respond
subsets are studied. to the questions presented.

R E V I E W Q UESTIONS
1. What is the most common clinical application of flow 2. Which of the following is true of CD45 antigen?
cytometry? a. It is present on every cell subpopulation in the bone
a. Diagnosis of platelet disorders marrow.
b. Detection of fetomaternal hemorrhage b. It is expressed on all hematopoietic cells with the
c. Diagnosis of leukemias and lymphomas exception of megakaryocytes and late erythroid
d. Differentiation of anemias precursors.
c. It is not measured routinely in flow cytometry.
d. It may be present on nonhematopoietic cells.
506 PART V Leukocyte Disorders

3. Erythroid precursors are characterized by the expression of: 7. Collection of ungated events:
a. CD71 a. Facilitates comprehensive analysis of all cells
b. CD20 b. Does not help in detection of unexpected abnormal
c. CD61 populations
d. CD3 c. Allows the collection of data on a large number of
rare cells
4. In the accompanying figure, the cell population colored in d. Is used for leukemia diagnosis only
aqua represents:
8. Mycosis fungoides is characterized by:
a. Loss of certain antigens compared with the normal
1023

T-cell population
b. Polyclonal T-cell receptor
c. Immunophenotype indistinguishable from that of
normal T cells
SS

d. Expression of CD3 and CD8 antigens

9. Mature granulocytes show the expression of:


a. CD15, CD33, and CD34
b. CD15, CD33, and CD41
0

100 101 102 103 c. CD15, CD33, and CD13


CD45-ECD d. CD15, CD33, and CD7

a. Monocytes 10. During the initial evaluation of flow cytometric data, cell
b. Nonhematopoietic cells size, cytoplasmic complexity, and expression of CD45
c. Granulocytes antigen are used to define cell subpopulations. Which of
d. Lymphocytes the following parameters defines cytoplasmic complexity/
granularity?
5. Antigens expressed by B-LL include: a. SS
a. CD3, CD4, and CD8 b. FS
b. CD19, CD34, and CD10 c. CD45
c. There are no antigens specific for B-LL. d. HLA-DR
d. Myeloperoxidase
11. The most important feature of the mature neoplastic B-cell
6. Which of the following is true of flow cytometric gating? population is:
a. It is best defined as selection of a target population a. The presence of a specific immunophenotype with
for flow cytometric analysis. expression of CD19 antigen
b. It can be done only at the time of data acquisition. b. A clonal light chain expression (i.e., exclusively - or
c. It can be done only at the time of final analysis and -positive population)
interpretation of flow cytometric data. c. A clonal T-cell receptor expression
d. It is accomplished by adjusting flow rate. d. Aberrant expression of CD5 antigen on CD19+ cells

REFERENCES
1. Swerdlow SH, Campo E, Harris NL, et al, editors: WHO 5. Human cell differentiation molecules. Available at: http://
classification of tumours of haematopoietic and lymphoid tissues, hcdm.org. Accessed May 28, 2010.
ed 4, Lyon, France, 2008, IARC Press. 6. Perfetto SP, Chattopadhyay PK, Roederer M: Seventeen-colour
2. Czader M, Ali SZ: Flow cytometry as an adjunct to cytomor- flow cytometry: unravelling the immune system. Nat Rev
phologic analysis of serous effusions. Diagn Cytopathol 29:74- Immunol 4:648-655, 2004.
78, 2003. 7. Stelzer GT, Shults KE, Loken MR: CD45 gating for routine
3. Wood BL, Arroz M, Barnett D, et al: 2006 Bethesda Interna- flow cytometric analysis of human bone marrow specimens.
tional Consensus recommendations on the immunophe Ann N Y Acad Sci 677:265-281, 1993.
notypic analysis of hematolymphoid neoplasia by flow 8. Borowitz MJ, Guenther KL, Shults KE, et al: Immunopheno-
cytometry: optimal reagents and reporting for the flow cyto- typing of acute leukemia by flow cytometric analysis: use of
metric diagnosis of hematopoietic neoplasia. Cytometry B Clin CD45 and right-angle light scattered to gate on leukemic
Cytom 72:S14-S22, 2007. blasts in three-color analysis. Am J Clin Pathol 100:534-540,
4. Khler G, Milstein C: Continuous cultures of fused cells 1993.
secreting antibody of predefined specificity. Nature 256:495- 9. Macedo A, Orfao A, Ciudad J, et al: Phenotypic analysis of
497, 1975. CD34 subpopulations in normal human bone marrow and
CHAPTER 33 Flow Cytometric Analysis in Hematologic Disorders 507
its application for the detection of minimal residual disease. 20. Kussick SJ, Wood BL: Four-color flow cytometry identifies
Leukemia 9:1896-1901, 1995. virtually all cytogenetically abnormal bone marrow samples
10. Terstappen LW, Safford M, Loken MR: Flow cytometric analy- in the workup of non-CML myeloproliferative disorders. Am
sis of human bone marrow: III. Neutrophil maturation. J Clin Pathol 120:854-865, 2003.
Leukemia 4:657-663, 1990. 21. Cazzola M: Flow cytometry immunophenotyping for diagno-
11. Loken MR, Shah VO, Dattilio KL, et al: Flow cytometric analy- sis of myelodysplastic syndrome. Haematologica 94:1041-
sis of human bone marrow: I. Normal erythroid develop- 1043, 2009.
ment. Blood 69:255-263, 1987. 22. Ogata K, Nakamura K, Yokose N, et al: Clinical significance
12. Ciudad J, Orfao A, Vidriales B, et al: Immunophenotypic of phenotypic features of blasts in patients with myelodys-
analysis of CD19+ precursors in normal human adult bone plastic syndrome. Blood 100:3887-3896, 2002.
marrow: implications for minimal residual disease detection. 23. Wells DA, Benesch M, Loken MR, et al: Myeloid and mono-
Haematologica 83:1069-1075, 1998. cytic dyspoiesis as determined by flow cytometric scoring in
13. Terstappen LW, Huang S, Picker LJ: Flow cytometric assess- myelodysplastic syndrome correlates with the IPSS and with
ment of human T-cell differentiation in thymus and bone outcome after hematopoietic stem cell transplantation. Blood
marrow. Blood 79:666-677, 1992. 102:394-403, 2003.
14. Andrieu V, Radford-Weiss I, Troussard X, et al: Molecular 24. Harbott J, Mancini M, Verellen-Dumoulin C, et al: Hemato-
detection of t(8;21)/AML1-ETO in AML M1/M2: correlation logical malignancies with a deletion of 11q23: cytogenetic
with cytogenetics, morphology and immunophenotype. Br J and clinical aspects. Leukemia 12:823-827, 1998.
Haematol 92:855-865, 1996. 25. Tabernero MD, Bortoluci AM, Alaejos I, et al: Adult precur-
15. Adriaansen H, Boekhorst PAW, Hagemeijer AM, et al: Acute sor B-ALL with BCR/ABL gene rearrangements displays a
myeloid leukemia M4 with bone marrow eosinophilia unique immunophenotype based on the pattern of CD10,
(M4Eo) and inv(16)(p13q22) exhibits a specific immuno- CD34, CD13 and CD38 expression. Leukemia 15:406-414,
phenotype with CD2 expression. Blood 81:3043-3051, 1993. 2001.
16. Lo Coco F, Avvisati G, Diverio D, et al: Rearrangements of the 26. Li S, Eshleman JR, Borowitz MJ: Lack of surface immuno-
RAR-alpha gene in acute promyelocytic leukaemia: correla- globulin light chain expression by flow cytometric immuno-
tions with morphology and immunophenotype. Br J Haema- phenotyping can help diagnose peripheral B-cell lymphoma.
tol 78:494-499, 1991. Am J Clin Pathol 118:229-234, 2002.
17. Krasinskas AM, Wasik MA, Kamoun M, et al: The usefulness 27. Beck RC, Stahl S, OKeefe CL, et al: Detection of mature T-cell
of CD64, other monocyte-associated antigens, and CD45 leukemias by flow cytometry using anti T-cell receptor V beta
gating in the subclassification of acute myeloid leukemias antibodies. Am J Clin Pathol 120:785-794, 2003.
with monocytic differentiation. Am J Clin Pathol 110:797- 28. Alaibac M, Pigozzi B, Belloni-Fortina A, et al: CD7 expression
805, 1998. in reactive and malignant human skin T-lymphocytes. Anti-
18. Helleberg C, Knudsen H, Hansen PB, et al: CD34+ mega- cancer Res 23:2707-2710, 2003.
karyoblastic leukaemic cells are CD38, but CD61+ and gly- 29. Richards SJ, Rawstron AC, Hillmen P: Application of flow
cophorin A+: improved criteria for diagnosis of AML-M7? cytometry to the diagnosis of paroxysmal nocturnal hemo-
Leukemia 11:830-834, 1997. globinuria. Cytometry 42:223-233, 2004.
19. Stetler-Stevenson M, Arthur DC, Jabbour N, et al: Diagnostic 30. Edwards BS, Oprea T, Prossnitz ER, et al: Flow cytometry for
utility of flow cytometric immunophenotyping in myelodys- high-throughput, high-content screening. Curr Opin Chem
plastic syndrome. Blood 98:979-987, 2001. Biol 8:392-398, 2004.
34 Myeloproliferative Neoplasms
Tim R. Randolph

OUTLINE OBJECTIVES
Chronic Myelogenous After completion of this chapter, the reader will be able to:
Leukemia
Incidence 1. Define myeloproliferative neoplasms (MPNs). 13. Discuss the progression of PV and treatment
Cytogenetics of the 2. List the most common diseases included in the clas- modalities.
Philadelphia sification of MPNs and recognize their abbreviations. 14. Define essential thrombocythemia (ET), with empha-
Chromosome 3. Define chronic myelogenous (chronic myelocytic) sis on the affected cell lines.
Molecular Genetics leukemia (CML), with emphasis on the affected cell 15. List the diagnostic criteria for ET.
Pathogenetic Mechanism lines. 16. Describe the morphologic changes in the peripheral
Peripheral Blood and 4. Discuss the pathogenic mechanism in CML, includ- blood in patients with ET.
Bone Marrow ing the cytogenetic and molecular biology. 17. Discuss two complications that may occur in patients
Other Laboratory 5. Describe the peripheral blood and bone marrow find- with ET.
Findings
ings in CML. 18. Define primary myelofibrosis (PMF), with emphasis
Progression
6. List the clinical phases of CML and describe the on the affected cell lines and pathogenesis.
Related Diseases
Treatment expected symptoms and laboratory test results in 19. Describe the key pathologic features of PMF in bone
Polycythemia Vera each phase. marrow, peripheral blood, and tissues.
Pathogenic Mechanism 7. Discuss the best approach to treatment and mecha- 20. Describe the course of disease of PMF and current
Diagnosis nisms of drug resistance. therapy.
Peripheral Blood and 8. Define polycythemia vera (PV), with emphasis on the 21. Briefly describe the other myeloproliferative
Bone Marrow affected cell lines. disorders.
Clinical Presentation 9. Discuss the JAK2 mutation and the proposed patho- 22. Given complete blood count and cytogenetic, molec-
Treatment and Prognosis genic mechanism in PV. ular, and other laboratory results, recognize the find-
Essential 10. Discuss clinical symptoms commonly observed in ings consistent with each major MPN.
Thrombocythemia
patients with PV. 23. Recommend follow-up testing for suspected MPN
Incidence
11. Identify major morphologic changes in the bone and interpret the results of testing.
Clinical Presentation
Diagnosis marrow and peripheral blood in patients with PV. 24. Briefly describe the other MPNs outlined in this
Peripheral Blood and 12. List diagnostic criteria for PV. chapter.
Bone Marrow
Treatment and Prognosis
CASE STUDY
Primary Myelofibrosis
Myelofibrosis After studying the material in this chapter, the reader should be able to respond to the following
Hematopoiesis and case study:
Extramedullary
Hematopoiesis A 34-year-old woman came to the physician with a 2-month history of increasing weakness, per-
Incidence and Clinical sistent nonproductive cough, fever and chills accompanied by night sweats, and a 13-lb weight loss
Presentation over a 6-month period. Results of chest radiographs and purified protein derivative test (for tuber-
Peripheral Blood and culosis) were negative. The patient was treated with ciprofloxacin and her cough improved, but she
Bone Marrow continued to grow weaker and was able to consume only small quantities of food. The patient
Immune Response appeared pale and cachectic. There was tenderness and fullness in the left upper quadrant, and the
Treatment and Prognosis spleen was palpable below the umbilicus. No hepatomegaly or peripheral adenopathy was noted.
Interconnection Her laboratory results were as follows:
Between Essential WBCs248 109/L
Thrombocythemia,
Hb9.5g/dL
Polycythemia Vera, and
Hct26.3%
Primary Myelofibrosis
Continued

508
CHAPTER 34 Myeloproliferative Neoplasms 509

OUTLINEcontd CASE STUDYcontd


Other Myeloproliferative After studying the material in this chapter, the reader should be able to respond to the following
Neoplasms case study:
Chronic Neutrophilic
Leukemia Platelets449 109/L
Chronic Eosinophilic Segmented neutrophils44%
Leukemia, Not Band neutrophils4%
Otherwise Specified Lymphocytes10%
Mastocytosis Eosinophils3%
Myeloproliferative Basophils7%
Neoplasm, Myelocytes30%
Unclassifiable Promyelocytes1%
Myeloblasts1%
Nucleated RBCs2 per 100WBCs
Reticulocytes3%
Leukocyte alkaline phosphatase (LAP) score20 (reference range, 40 to 130)
Lactate dehydrogenase692IU (reference range, 140 to 280IU)
Uric acid8.1mg/dL (reference range, 4 to 6mg/dL)
1. What is the significance of the elevated WBC count and abnormal WBC differential?
2. How does the LAP score aid in the diagnosis?
3. Justify the use of cytogenetic studies in a patient with test results similar to those in this case
study.
4. Predict the results of the cytogenetic studies.
5. Describe the molecular mutation resulting from the cytogenetic abnormality.
6. What is the usual treatment for this disorder?
7. Briefly discuss mechanisms of drug resistance.

T
he myeloproliferative neoplasms (MPNs) are clonal ET is characterized by increased megakaryocytopoiesis and
hematopoietic disorders caused by genetic mutations in peripheral blood thrombocytosis.10
the hematopoietic stem cells that result in expansion, MPNs present as stable chronic disorders that may trans-
excessive production, and overaccumulation of erythrocytes, form first to a subacute, then to an aggressive cellular growth
granulocytes, and platelets. Expansion occurs in varying com- phase, such as acute myeloid leukemia (AML) or acute lym-
binations in the bone marrow, peripheral blood, and tissues.1-4 phoblastic leukemia (ALL). They may manifest a depleted cel-
The MPNs have pathogenetic similarities as well as common lular phase such as bone marrow hypoplasia, or exhibit clinical
clinical and laboratory features.5 symptoms and morphologic patterns characteristic of first a
MPNs are predominately chronic with accelerated, sub- subacute, then a more aggressive cellular expression.
acute, or acute phases. In certain patients it is difficult to make Familial MPNs have been described in families in which
a clear delineation between subacute and chronic phases from two or more members are affected.11
clinical and morphologic findings.
The World Health Organization (WHO) has classified the
CHRONIC MYELOGENOUS LEUKEMIA
MPNs as including four predominant disorders: chronic
myelogenous leukemia (CML); polycythemia vera (PV) also Chronic myelogenous leukemia (CML) is an MPN arising from
known as polycythemia rubra vera; essential (primary) thrombo- a single genetic translocation in a pluripotential hematopoietic
cythemia (ET); and primary myelofibrosis (PMF), also known stem cell producing a clonal overproduction of the myeloid
as agnogenic myelofibrosis with myeloid metaplasia and chronic cell line, which results in a preponderance of immature cells
idiopathic myelofibrosis. Several other less common MPN condi- in the neutrophilic line. CML begins with a chronic clinical
tions have been described and are classified as chronic neutro- phase that progresses to an accelerated phase in 3 to 4 years
philic leukemia (CNL); chronic eosinophilic leukemia (CEL), and often terminates as an acute leukemia if left untreated. The
not otherwise specified; mastocytosis; and myeloproliferative clinical features are frequent infection, anemia, bleeding, and
disorder, unclassified.6 CML and PV are defined by their over- splenomegaly, all secondary to massive pathologic accumula-
production of granulocytes and erythrocytes, respectively.2,7,8 tions of myeloid progenitor cells in bone marrow, peripheral
PMF is a combination of overproduction of hematopoietic cells blood, and extramedullary tissues. Neutrophilia with all matu-
and stimulation of fibroblast production leading to ineffective rational stages present, basophilia, eosinophilia, and often
hematopoiesis with resultant peripheral blood cytopenias.9 thrombocytosis are noted in peripheral blood. The clonal
510 PART V Leukocyte Disorders

origin of hematopoietic cells in CML has been verified gene is approximately 100 kb with 20 exons. In 1984 Groffen
in studies of females heterozygous for glucose-6-phosphate et al identified the BCR on chromosome 22 as a 5-exon region
dehydrogenase. Only one isoenzyme is active in affected cells, involving exons 12 to 16 that was the area of breakage in the
whereas two isoenzymes are active in nonaffected cells.12 traditional t(9;22) translocation.19 This area was later termed
the major BCR. Two other areas of breakage were identified on
Incidence chromosome 22, one nearer the 5 (head) of the BCR1 gene,
CML occurs at all ages but is seen predominantly in those aged called the minor BCR, and one in the 3 end (tail) of the BCR1
46 to 53 years. It represents about 20% of all cases of leukemia, gene termed the micro BCR.
is slightly more common in males than in females, and carried Within the major BCR two specific breakpoints account for
a mortality rate of 1.5 per 100,000 per year in the era before the t(9;22) translocation involved in the development of CML.
the development of imatinib mesylate (Gleevec). Imatinib is a Therefore, two areas of breakage in the major BCR combined
tyrosine kinase inhibitor that has changed the prognosis and with one in the minor BCR and one in the micro BCR produce
treatment for CML and is described in detail later. the four versions of the BCR/ABL1 chimeric gene. Because the
Symptoms associated with clinical onset are usually of two breakpoints in the major BCR differ by only one exon, the
minimal intensity and include fatigue, decreased tolerance of chimeric protein product is essentially the same size and is
exertion, anorexia, abdominal discomfort, weight loss, and designated as the p210 protein. Breakage in the BCR1 gene in
symptomatic effects from splenic enlargement. the major BCR contributes exons 1 to 13 or 1 to 14, whereas
the ABL1 gene contributes exons 2 to 11. Breakage in the minor
Cytogenetics of the Philadelphia Chromosome BCR contributes only exon one from BCR, which joins with
A unique chromosome, the Philadelphia chromosome, is the same exons 2 to 11 of ABL1 to produce a p190 protein.
present in proliferating hematopoietic stem cells and their The micro BCR breakpoint contributes exons 2 to 19 from
progeny in CML and must be identified to confirm the diag- BCR1, which fuse with ABL1 exons 2 to 11, producing the p230
nosis. Although the cause of Philadelphia chromosome forma- protein. Therefore, the four possible BCR1 breakpoints produce
tion is unknown, it appears more frequently in populations four different chimeric genes, resulting in a total of three dif-
exposed to ionizing radiation.13,14 In most patients, a cause ferent protein products.20
cannot be identified. Appearance of the Philadelphia chromo-
some in donor cells after allogeneic bone marrow trans Pathogenetic Mechanism
plantation indicates the possibility of a transmissible agent.15 To understand the aberrant function of the BCR/ABL fusion
The Philadelphia chromosome was first identified as a short protein, it is first helpful to understand both the normal BCR
chromosome 22 in 1960 by Nowell and Hungerford in and ABL proteins. The wild-type ABL protein, when in its usual
Philadelphia.16 In 1973 Rowley, of the University of Illinois at location on chromosome 9, codes for p125, which exhibits
Chicago, discovered that the Philadelphia chromosome is a normal tyrosine kinase activity. The BCR1 gene produces p160,
reciprocal translocation between the long arms of chromo- expresses serine and threonine kinase activity, and is thought
somes 9 and 22 (see Chapter 31).17 This acquired somatic to function in the regulation of cell growth. Protein kinases are
mutation specifically reflects the translocation of an ABL proto- enzymes that catalyze the transfer of phosphate groups from
oncogene from band q34 of chromosome 9 to the breakpoint adenosine triphosphate (ATP), guanosine triphosphate, and
cluster region (BCR) of band q11 of chromosome 22, which other phosphate donors to receiver proteins. A tyrosine kinase
results in a unique chimeric gene, BCR/ABL.18 This new gene transfers the phosphate group to a tyrosine amino acid on the
produces a 210-kD BCR/ABL fusion protein (p210BCR/ABL) that receiver protein. For the kinase activity of the ABL protein to
expresses enhanced tyrosine kinase activity compared with its occur, the ABL protein must first be phosphorylated. This is
natural enzymatic counterpart. often accomplished through autophosphorylation. The ABL
protein has three primary domains called SR1, SR2, and SR3
Molecular Genetics that together express and regulate the kinase activity. SR1 is the
The t(9;22) translocation that produces the BCR/ABL1 chimeric binding site for ATP; SR2 is the docking point for phosphate
gene has been observed in four primary molecular forms that receiver proteins; and SR3 is the domain that controls the
produce three versions of the BCR/ABL chimeric protein: p190, phosphorylation activity. When ATP binds to the ATP binding
p210, and p230 (Figure 34-1). The four genetic variations are site, the phosphate is transferred to the SR2 region of the ABL
based on the area of the BCR gene that houses the breakpoint protein, which initiates a conformational change that alters the
on chromosome 22, because the breakpoint on chromosome tertiary structure of the protein and exposes the active site of
9 occurs in the same location. The wild-type (normal) ABL1 the enzyme. When a second ATP binds the ATP binding site
gene on chromosome 9 is a relatively large gene of approxi- and a receiver protein docks in the SR2 domain, the phosphate
mately 230 kilobases (kb) containing 11 exons. The breakpoint group is transferred to the receiver protein. In most physiologi-
consistently occurs 5 of the second exon such that exons 2 to cally normal intracellular pathways, protein phosphorylation
11 are contributed to the BCR/ABL1 fusion gene. activates the receiver proteins (Figure 34-2). This phosphoryla-
There are four BCR genes in the human genome: BCR1, tion initiates a cascade of phosphorylation events, each activat-
BCR2, BCR3, and BCR4. It is the BCR1 gene that is involved ing the next protein until a transcription factor becomes
in the Philadelphia translocation. Wild-type (normal) BCR1 activated. These activation cascades, called signal transduction
CHAPTER 34 Myeloproliferative Neoplasms 511

Panel A
Chromosome 22 Chromosome 9
Normal BCR1 Gene Normal ABL Gene
Exons
1 1 2 3 4 5 6 7 8 9 10 11 1 2 3 4 5 17 18 19 20 1 1 2 3 4 5 6 7 8 9 10 11

3 1 2 4
minor major micro
bcr bcr bcr
5 End (Head) of BCR  3 End (Tail) of ABL
Four Common Fusion Genes Produce Three Fusion Proteins

Panel B
Two Most Common Fusion Genes Involve the Major BCR and Form One Fusion Protein

Exons 1 1 2 3 4 5 6 7 8 9 10 11 1 2 2 3 4 5 5 7 8 9 10 11
1
p210 bcrabl
Exons 1 1 2 3 4 5 6 7 8 9 10 11 1 2 3 2 3 4 5 5 7 8 9 10 11
2

Panel C
Less Common Fusion Gene Involves the Minor BCR and Forms One Fusion Protein

Exons 1 1 2 3 4 5 6 7 8 9 10 11
3 p 190 bcrabl

Panel D
Least Common Fusion Gene Involves the Micro BCR and Forms One Fusion Protein
Exons 1 1 2 3 4 5 6 7 8 9 10 11 1 2 3 4 5 17 18 19 2 3 4 5 5 7 8 9 10 11
p230
4 bcrabl

bcr  breakpoint cluster region, abl  Abelson oncogene

Figure 34-1 Molecular biology of the BCR/ABL fusion gene. A, Normal BCR1 gene on chromosome 22 and ABL gene on chromosome 9. B, Two BCR
fusion gene products from the major BCR. C, Fusion gene product from the minor BCR. D, Fusion gene product from the micro BCR.

pathways, are designed to activate genes necessary to control proliferation from activation of the RAS gene and a decrease
cell proliferation, differentiation, and natural cell death, called in or resistance to apoptosis. New clones of stem cells vulner-
apoptosis. There are several signal transduction pathways acti- able to additional genetic changes lead to the accelerated and
vated by the ABL tyrosine kinase that function in concert to blast phases of CML. In addition, the BCR/ABL protein local-
activate these genes in a precise order and at the required level izes in the cytoplasm, rather than in the nucleus as does the
of activation to control these cellular events.20,21 normal ABL protein. The mutation affects maturation and dif-
In the case of CML, the BCR/ABL1 translocation occurs next ferentiation of hematopoietic and lymphopoietic cells, whose
to the SR3 domain of the ABL1 moiety, which is designed to progeny eventually dominate in the affected individual.
control the rate and timing of phosphorylation. Therefore, the Progeny cells that exhibit this chromosome include neutro-
BCR/ABL tyrosine kinase has lost the ability to shut off kinase phils, eosinophils, basophils, monocytes, nucleated erythro-
activity and is said to have constitutive tyrosine kinase activity. cytes, megakaryocytes, and B lymphocytes.7,23
The BCR/ABL enzyme continuously adds phosphate groups to In addition, the loss of genetic segments in the 5 end of the
tyrosine residues on cytoplasmic proteins, activating several ABL1 gene results in an altered protein-binding affinity for
signal transduction pathways. These pathways stimulate gene F-actin, which leads to a reduction in contact binding of hema-
expression keeping the myeloid cells proliferating, reducing topoietic CML cells to stromal cells and causing premature
differentiation, reducing adhesion of cells to bone marrow release of cells into the circulation.21 Abnormal adhesion
stroma, and virtually eliminating apoptosis. The result is between stem cells and stroma may dysregulate hematopoiesis.
increased clonal proliferation of myeloid cells secondary to One action of interferon- therapy is to reverse the loss of
a reduction in or loss of sensitivity to protein regulators.22 adhesion of CML progenitor cells, which reduces the prema-
There is an increase in growth factorindependent cellular ture release of these cells into the circulation.24
512 PART V Leukocyte Disorders

Extracellular space

Cytoplasm

Coiled Serine GDP-GTP Binding Sites


Coil Threonine Exchange sh 2 Nucleus, DNA, Actin
DD Y177 Kinase Factor sh 3 sh 1

BAP-1 CBL CRKL Adaptor


Proteins
GRB2 SHC
Signal Transduction Pathways
Adhesion
Transformation Proliferation Apoptosis SFK
FAK Actin
PI-3
SOS JAK-STAT
Kinase
Nuclear
p85 p110
AKT Binding
Proteins
RAS-GTP MAPK
Nucleus

Figure 34-2 Signal transduction pathways influenced by the BCR/ABL fusion protein.

Apoptotic functions are lost because the BCR/ABL fusion myelocytes predominate, and immature and mature eosino-
protein has a propensity to be sequestered in the cytoplasm, phils and basophils are increased. Myeloblasts and promyelo-
which has antiapoptotic functions. The p210 is necessary for cytes are present at a rate of approximately 1% and 5%,
CML transformation of the hematopoietic stem cell. respectively. Lymphocytes and monocytes are present and
The BCR/ABL1 fusion is also identified with Philadelphia often show an absolute increase in number but a relative
chromosomepositive ALL. The chromosome appears in 20% decrease in percentage. Nucleated red blood cells (NRBCs) are
of adults and 2% to 5% of children with this disease. The rare. Platelets are normal or increased, and some may exhibit
minor chimeric BCL/ABL1 gene that transcribes and translates abnormal morphology.
to a p185/p190 protein is present in 50% of Philadelphia Bone marrow changes are illustrated in Figure 34-4. An
chromosomepositive ALL cases in adults and 75% of intense hypercellularity is present due to granulopoiesis,
Philadelphia chromosomepositive ALL cases in children. The marked by broad zones of immature granulocytes, usually peri-
micro BCR, when fused with the ABL1 gene, produces a large vascular or periosteal, differentiating into more centrally placed
p230 protein that is the least common version found. mature granulocytes. Normoblasts appear reduced in number.
Megakaryocytes are normal or increased in number and, when
Peripheral Blood and Bone Marrow increased, may appear in clusters and exhibit dyspoietic cyto-
There are dramatic morphologic changes in the peripheral logic changes. They often appear small with reduced nuclear
blood and bone marrow that reflect the expansion of the gran- size (by approximately 20%) and reduced nuclear lobulations.
ulocyte pool, particularly in the later maturational stages. Table Reticulin fibers are increased in approximately 20% of patients.
34-1 lists the qualitative changes in the peripheral blood, bone Increased megakaryocyte density is associated with an increase
marrow, and extramedullary tissues that are commonly in myelofibrosis.25 The presence of pseudo-Gaucher cells (see
observed at the time of diagnosis. A dramatic left shift is noted Chapter 28) usually occurs.
that extends down to the promyelocyte stage and occasionally
even produces a few blasts in the peripheral blood. The platelet Other Laboratory Findings
count is often elevated, reflecting the myeloproliferative nature Hyperuricemia and uricosuria from increased cell turnover
of the disease. Extramedullary granulopoiesis may involve may be associated with secondary gout, urinary uric acid
sinusoids and medullary cords in the spleen and sinusoids, stones, and uric acid nephropathy.26 Approximately 15% of
portal tract zones, and solid areas of the liver. patients exhibit total white blood cell (WBC) counts greater
Figure 34-3 illustrates a common pattern in the peripheral than 300 109/L.27 Symptoms in these patients are secondary
blood film of chronic phase CML at the time of diagnosis. to vascular stasis and possible intravascular consumption of
Leukocytosis is readily apparent at scanning microscopic oxygen by the leukocytes. Symptoms are reversible with the
powers. Segmented neutrophils, bands, metamyelocytes, and lowering of the total WBC count.28
CHAPTER 34 Myeloproliferative Neoplasms 513

TABLE 34-1 Common Morphologic Changes in Chronic Blast crisis involves the peripheral blood, bone marrow, and
Myelogenous Leukemia extramedullary tissues. By definition, blasts constitute more
than 20% of total bone marrow cellularity; the peripheral
Peripheral Blood blood exhibits increased blasts.29 Blast crisis leukemia usually
Erythrocytes Normal or decreased
is AML or ALL, but origins from other hematopoietic clonal
Reticulocytes Normal
cells are possible. Extramedullary growth may occur as lym-
Nucleated red blood cells Present
phocytic or myelogenous cell proliferations; the latter are often
Total white blood cells Increased
referred to as granulocytic sarcoma. Extramedullary sarcoma is
Lymphocytes Normal or increased
observed at many sites or locations in the body and may
Neutrophils Increased
precede a marrow blast crisis. The clinical symptoms of blast
Basophils Increased
crisis mimic those of acute leukemia, including severe anemia,
Eosinophils Increased
leukopenia of all WBCs except blasts, and thrombocytopenia.
Myelocytes Increased
Chromosome abnormalities such as additional Philadelphia
Leukocyte alkaline phosphatase Decreased
chromosome(s), isochromosome 17, trisomy 8, loss of Y chro-
Platelets Normal or increased
mosome, and trisomy 19 accumulate with disease progres-
sion.31,32 These generally occurred in approximately 75% of
Bone Marrow patients in the pre-imatinib era.
Cellularity Increased
Granulopoiesis Increased
Related Diseases
Erythropoiesis Decreased
Several diseases exist that are clinically similar to CML but do
Megakaryopoiesis Increased or normal
not exhibit the Philadelphia chromosome and express only a
Reticulin Increased
few pseudo-Gaucher cells. Chronic neutrophilic leukemia is
Macrophages
another MPN that manifests with peripheral blood, bone
Gaucherlike Sea blue
marrow, and extramedullary infiltrative patterns similar to
Green-gray crystals Increased
those of CML, except that only neutrophilic granulocytes are
Megakaryocytes
present and fewer than 10% of peripheral blood neutrophils
Small Increased
are immature.33 Similarly, chronic monocytic leukemia involves
a comparable expansion of monocytes, including functional
Extramedullary Tissue monocytes.34
Splenomegaly Present
Juvenile myelomonocytic leukemia and adult chronic
Sinusoidal Present
myelomonocytic leukemia are classified by the WHO as
Medullary Present
myelodysplastic/myeloproliferative diseases because of the
Hepatomegaly Present
overlap in clinical, laboratory, or morphologic findings. Juve-
Sinusoidal Present
nile myelomonocytic leukemia is observed in children younger
Portal tract Present
than 4 years of age and is accompanied by an expansion in the
Local infiltrates Present
number of monocytes and granulocytes, including immature
granulocytes, and manifestations of dyserythropoiesis.35
The peripheral blood of adults with chronic myelomono-
cytic leukemia may have characteristics similar to those seen
Progression in the refractory anemias, such as oval macrocytes and reticu-
In the pre-imatinib era, most patients disease would eventually locytopenia. The peripheral WBC concentration may reach
transform into acute leukemia.29 Before blastic transformation, 100 109/L. According to WHO criteria, absolute monocytosis
some patients proceed through an intermediate metamorphosis (more than 1 109 monocytes/L) must be present to make the
or accelerated phase. Disease progression is accompanied by an diagnosis. Clinical features include prominent splenomegaly,
increase in the frequency and number of clinical symptoms, symptoms of anemia, fever, bleeding, and infection. Before
adverse changes in laboratory values, and poorer response to presence of the Philadelphia chromosome was established as
therapy than in the chronic phase. Additional chromosome a requirement for the diagnosis of CML, some cases that were
abnormalities may appear, associated with enhanced dyshema- classified as Philadelphia chromosomenegative CML likely
topoietic cell maturation patterns and increases in morphologic represented misdiagnoses of chronic myelomonocytic leuke-
and functional abnormalities in blood cells. There is often an mia.36 Chronic myelomonocytic leukemia is discussed further
increasing degree of anemia and, in the peripheral blood, fewer with myelodysplastic syndromes in Chapter 35.
mature leukocytes, more basophils, and fewer platelets, with a A puzzling group of patients exhibit Philadelphia
greater proportion of abnormal platelets, micromegakaryo- chromosomepositive acute leukemia. Studies reveal that 2%
cytes, and megakaryocytic fragments. The circulating blast of patients with AML exhibit Philadelphia chromosome in a
count increases to 10% to 19%. This total blast percentage, or significant proportion of blasts. Further, 5% of patients with
a combination of 20% blasts and promyelocytes, has been childhood-onset ALL and 20% of those with adult-onset ALL
proposed as a diagnostic criterion for the accelerated phase.30 test positive for Philadelphia chromosome.37-40 The proper
514 PART V Leukocyte Disorders

A B

C
Figure 34-3 Peripheral blood films in the chronic phase of chronic myelogenous leukemia. A, Leukocytosis is evident at scanning power (100). B, Bimodal
population of segmented neutrophils and myelocytes (500). C, Increased basophils and immature neutrophils (1000).

a late-stage mutation that contributed little to acute leukemia


leukemogenesis.

Treatment
Early treatment approaches for CML were unable to produce
remission, so the goal of therapy became the reduction of
tumor burden. The first forms of therapy for CML included
alkylating agents such as nitrogen mustard,41 introduced in the
late 1940s, and busulfan,42 which came into use in the early
1950s. Later, busulfan in combination with 6-thioguanine was
used to achieve the goal of tumor burden reduction. Other
drugs like hydroxyurea and 6-mercaptopurine were introduced
later and found to improve patient survival. The discovery
of interferon- in 1983 dramatically improved outcomes
Figure 34-4 Bone marrow biopsy specimen in the chronic phase of of patients with CML by inducing the suppression of the
chronic myelogenous leukemia, showing hypercellularity with increased Philadelphia chromosome, reducing the rate of cellular pro-
granulocytes and megakaryocytes (hematoxylin and eosin stain, 400). gression to blast cells, and increasing the frequency of long-
term patient survival.43
Interferon- stimulates a cell-mediated antitumor host
alignment of these cases within the spectrum of CML is specu- response that reduces myeloid cell numbers, induces cytoge-
lative. It is understood that some of these cases likely represent netic remissions, and increases survival.44 It improves the fre-
undiagnosed CML that rapidly progressed to an acute leukemia quency and duration of hematologic remission and reduces the
prior to diagnosis. However, because rapidly dividing malig- frequency of detection of the Philadelphia chromosome. In
nant cells are more prone to genetic mutation, the presence of some patients, a complete cytogenetic remission is achieved for
the Philadelphia chromosome in acute leukemias may reflect a time.
CHAPTER 34 Myeloproliferative Neoplasms 515

Panel A
BCR-ABL BCR-ABL BCR-ABL BCR-ABL
Unphosphorylated Phosphorylated Bound ABL Bound ABL
No kinase activity Kinase activity Functional state Phosphorylated

ATP
SH1 SH1 SH1 ATP SH1 ATP
PO4 PO4
PO4 PO4 PO4
Receiver Receiver Receiver
SH2 SH2 SH2 Protein SH2 Protein Protein
Receiver Activates
Protein signal
SH3 SH3 SH3 SH3 transduction
pathways

Panel B
BCR-ABL BCR-ABL
Unphosphorylated Bound ABL
No kinase activity ATP Stable closed form

ATP
SH1 Gleevec SH1 Gleevec

Receiver
SH2 SH2 Protein

Receiver Receiver No activation


Protein Protein of signal
SH3 SH3 transduction
pathways

Figure 34-5 Mechanism of imatinib mesylate inhibition of BCR/ABL tyrosine kinase activity. A, Mechanism of tyrosine kinase activity of the BCR/ABL fusion
protein. B, Mechanism of tyrosine kinase inhibition by imatinib mesylate.

In 1997 it was discovered that cytarabine given with protein. When imatinib binds the ATP binding site, ATP is
interferon- improved the frequency of hematologic remis- unable to bind to provide the phosphate group necessary for
sions but did not eliminate the BCR/ABL gene, which was still kinase activity. Imatinib binds the BCR/ABL protein in the
detected by molecular and fluorescent methods.45 Also, in inactive conformation, which precedes the autophosphoryla-
some patients the side effects of therapy became severe, drug tion necessary to generate the kinase active site (Figure 34-5).47
resistance appeared, and relapse rates were not improved com- Goals of therapy include complete hematologic remission
pared with other chemotherapies. and cytogenetic remission induced in part by the reactivation
Bone marrow and stem cell transplantation with either of apoptotic pathways.48 The effectiveness of imatinib therapy
autologous or allogeneic hematopoietic stem cells have been and stem cell transplantation is best monitored using quantita-
reported as curative, especially in patients younger than age 55. tive real-time reverse transcriptase polymerase chain reaction
Relapses occur, but long-term, disease-free survival is possible. (RT-PCR). These monitoring tools are used to determine com-
Optimal survival occurs when the patient is treated during the plete cytogenetic and molecular remission. The most sensitive
chronic phase within 1 year of diagnosis and is younger than measure of the effectiveness of imatinib therapy is the number
age 50. Treatment requires ablative chemotherapy followed by of log reductions of BCR/ABL transcripts using real-time
transplantation of mobilized normal progenitor cells that RT-PCR.49 Expected therapeutic response to imatinib is com-
exhibit CD34+ surface markers. Allogeneic bone marrow trans- plete hematologic remission in 3 to 6 months, complete cyto-
plants are more successful in patients up to age 55 when genetic response in 6 months to 1 year, and a 2 to 3 log
donors are matched for HLA antigens A, B, and DR. Donor- reduction in BCR/ABL transcripts. When real-time RT-PCR is
matched lymphocyte infusions after allogeneic transplantation used, the greatest log reduction possible is a more than 4 log
of marrow from a sibling donor may assist in producing com- reduction, which represents the maximum sensitivity of the
plete remissions.46 assay. However, discontinuation of imatinib therapy in patients
Modern therapies involve the use of synthetic proteins that who achieve a more than 4 log reduction will result in relapse.
bind the abnormal BCR/ABL protein, blocking the constitutive Although imatinib has proven to be a successful form of
tyrosine kinase activity and reducing signal transduction activa- therapy, a major limitation is the development of imatinib
tion. Imatinib mesylate is a synthetic tyrosine kinase inhibitor resistance resulting in relapse. Approximately 4% of patients
designed to selectively bind the ATP binding site and thus with newly diagnosed CML will become imatinib resistant.
inhibit the tyrosine kinase activity of the BCR/ABL fusion There are two major categories of imatinib resistance, primary
516 PART V Leukocyte Disorders

BCR-ABL BCR-ABL BCR-ABL


Unbound ABL Bound ABL Bound ABL
Resting state Functional state Phosphorylated

ATP
SH1 SH1 ATP SH1 ATP
PO4 PO4
PO4 PO4 PO4
Receiver Receiver Receiver
SH2 SH2 Protein SH2 Protein Protein

Receiver Activates
Protein signal
SH3 SH3 SH3 transduction
pathways

ATP
SH1 SH1 ATP SH1 ATP
PO4 PO4
PO4 PO4 PO4
Receiver Receiver Receiver
SH2 SH2 Protein SH2 Protein Protein

Receiver Activates
Protein signal
SH3 SH3 SH3 transduction
pathways

ATP
SH1 SH1 ATP SH1 ATP
PO4 PO4
PO4 PO4 PO4
Receiver Receiver Receiver
SH2 SH2 Protein SH2 Protein Protein

Receiver Activates
Protein signal
SH3 SH3 SH3 transduction
pathways

Figure 34-6 Mechanism of imatinib mesylate resistance due to an increased copy number of BCR/ABL genes. The increased copies produce more
BCR/ABL fusion proteins, which results in an increased tyrosine kinase activity requiring a higher dosage of imatinib to restore remission.

and secondary. Primary resistance is defined as the inability to mutations. Mutations in the ATP binding site reduce the
reach the remission milestones. This form of resistance is rare binding affinity of imatinib, producing some level of resistance
and probably results from the presence of mutations other (Figure 34-7). Second- and third-generation tyrosine kinase
than the BCR/ABL mutation at the time of diagnosis. Secondary inhibitors like dasatinib (Sprycel), nilotinib (Dasigna), and
resistance involves the loss of a previous response and occurs bosutinib (still in clinical trials at the time of publication)
at a rate of 16% at 42 months. The majority of cases of imatinib overcome the ATP binding site mutations except the T315I
resistance result from two primary causes: acquisition of addi- mutation. However, drugs designed to bind the A loop (receiver
tional BCR/ABL mutations and expression of point mutations protein binding site) will inhibit tyrosine kinase activity and
in the ATP binding site. Additional BCR/ABL mutations can overcome the T315I mutation.50,51 Currently, studies are under
occur through the usual translocation of the remaining unaf- way to evaluate modification of the dosage of imatinib used,
fected chromosomes 9 and 22, which converts the hematopoi- to identify and develop other tyrosine kinase inhibitors, and
etic stem cell from heterozygous to homozygous for the BCR/ to discover new classes of inhibitors that may be more effective
ABL mutation. A double dose of BCR/ABL can also be acquired than currently known tyrosine kinase inhibitors.
from gene duplication during mitosis and accounts for 10% The development of a care plan for treating a patient with
of secondary mutations. An additional BCR/ABL mutation newly diagnosed CML requires not only the formulation of
will double the tyrosine kinase activity, making the imatinib alternative approaches to achieve complete cellular remission,
dosage inadequate. In these cases higher dosages of imatinib but also the establishment of parameters to follow that confirm
will restore remission in most patients (Figure 34-6). The long-term success of therapy. Previous chemotherapies have
majority of patients who do not respond to higher dosages of provided cellular remission but usually do not suppress or
imatinib express point mutations in the ATP binding site. Over delay clinical transitions to accelerated or blast phases. Bone
50 mutations have been identified in the ATP binding site, and marrow transplantation for patients who qualify is likely the
these account for the remaining 50% to 90% of secondary preferred choice, but the long-term success (cure) rate remains
CHAPTER 34 Myeloproliferative Neoplasms 517

BCR-ABL BCR-ABL BCR-ABL BCR-ABL


Mutated ATP Binding Site Auto-phosphorylated Bound ATP Bound Receiver Protein
Reduced Binding Increased Kinase Activity Functional State Phosphorylates Protein
Affinity of Gleevec ATP
ATP
SH1 Gleevec SH1 Gleevec SH1 ATP SH1 ATP
PO4 PO4
PO4 PO4 PO4
Receiver Receiver Receiver
SH2 SH2 SH2 SH2 Protein Protein
Protein

Receiver Receiver Activates Protein


Protein Protein Continuous
SH3 SH3 SH3 SH3 Stimulation of
Signal Transduction
Pathway

Figure 34-7 Mechanism of imatinib mesylate resistance due to point mutations in the adenosine triphosphate (ATP) binding site. The mutations reduce
the binding affinity of imatinib, allowing ATP to bind; this restores the increased tyrosine kinase activity that drives the phenotype and pathogenesis of the
disease.

at 50% to 70%. For a patient to qualify for transplantation, the some level of normal hematopoiesis.53 Adverse clinical
patient must be younger than 50 years of age, must be in progression seems to correlate with the propagation of the
the first year of the disease, and must have CML that is still in erythropoietin-sensitive colony-forming units.54 Understand-
the chronic phase, and a histocompatible donor must be avail- ing of the pathologic mechanism explaining this phenomenon
able. Today, imatinib is considered first-line therapy for all in PV was significantly advanced in 2005 with the discovery of
patients with newly diagnosed CML. For the small subset of a consistent mutation in the JAK2 gene. The specific JAK2 muta-
patients who qualify for hematopoietic stem cell transplanta- tion, JAK2 V617F, is detected in 90% to 97% of patients with
tion, imatinib is used to induce hematologic remission prior PV. Shortly after the JAK2 V617F mutation was reported, several
to transplantation. For all other CML patients, imatinib is used groups corroborated the finding using other approaches and
as first-line therapy unless remission is not achieved (primary showed that the mutation is acquired, clonal, present in the
resistance) or until relapse occurs following remission (second- hematopoietic stem cell, constitutively active, and capable of
ary resistance). Once the cause of relapse has been determined activating the erythropoietic signal transduction pathway in the
by cytogenetic and molecular testing, either a higher dosage of absence of erythropoietin. The point mutation changes the
imatinib can be given (for an additional BCR/ABL mutation) amino acid at position 617 from valine to phenylalanine,
or a second- or third-generation tyrosine kinase inhibitor which unlocks the inhibition of the tyrosine kinase and con-
(dasatinib, nilotinib, bosatinib) can be prescribed unless the verts it to the active conformation. More specifically, the phe-
mutation is the T315I mutation in the ATP binding site. If the nylalanine mutation in the kinase domain is unable to bind
T315I mutation is detected, the patient can be given an A-loop the corresponding amino acid in the pseudokinase domain, as
inhibitor (ONO12380) or other drugs like MK 0457 or BIRB- can the wild-type counterpart valine, which prevents the protein
796 that inhibit the T315I mutation.51 from folding and remaining inactive (Figure 34-8). Normally,
erythropoietin is released from the kidney into the blood in
response to hypoxia and binds to erythropoietin receptors on
POLYCYTHEMIA VERA
the surface of erythroid precursor cells. The resulting confor-
Polycythemia vera (PV) is a neoplastic clonal myeloprolifera- mational change in the erythropoietin receptor causes two
tive disorder that commonly manifests with panmyelosis in the erythropoietin receptors to dimerize. This produces a docking
bone marrow and increases in erythrocytes, granulocytes, and point for inactive JAK2, which becomes activated through phos-
platelets in the peripheral blood.2 Splenomegaly is common. phorylation. Phosphorylated JAK2 undergoes a conformational
The disease arises in a hematopoietic stem cell. The hypothesis change, and the valine releases from the pseudokinase domain,
of a clonal origin for PV is supported by studies of X-linked which converts it to an active tyrosine kinase. JAK2 phosphory-
restriction fragment length deoxyribonucleic acid (DNA) lates several cytoplasmic proteins, but the signal transducer and
polymorphisms that demonstrate monoclonal X chromosome activator of transcription (STAT) proteins are the main target.
inactivation in all blood cells.52 A cascade of phosphorylations produce activated transcription
factors that activate a host of genes designed to drive and
Pathogenic Mechanism control cell proliferation and differentiation while also initiat-
In PV, neoplastic clonal stem cells are hypersensitive to, or ing apoptosis (Figure 34-9). Constitutive tyrosine kinase activ-
function independently of, erythropoietin for cell growth. Trace ity of the JAK2 protein causes continuous activation of several
levels of erythropoietin in serum stimulate the growth of ery- signal transduction pathways that are normally activated
throid progenitor cells in in vitro colony-forming growth following erythropoietin stimulation via the erythropoietic
systems. There is preservation of hypersensitive and normosen- receptor. Active JAK2 will phosphorylate STAT proteins in the
sitive erythroid colony-forming units, however, which indicates absence of erythropoietin or will overphosphorylate in its
518 PART V Leukocyte Disorders

Panel A Inactive JAK2

JH7 JH6 JH5 JH4 JH3 JH2


JH1

SOCS1
JAB

Active JAK2 P P

JH7 JH6 JH5 JH4 JH3 JH2 JH1 P

P P
SOCSI = Suppressor of cytokine signalling
JAB = Janus binding protein
Panel B Abnormal JAK2 Gene

N JH7 JH6 JH5 JH4 JH3 JH2 JH1


FERM SH2-like Pseudo Kinase
kinase
V617

JH7 JH6 JH5 JH4 JH3 JH2 JH1


Abnormal JAK2 protein

Activation of pseudokinase domain of JAK2 protein

Constitutive kinase activity

Stimulation of STAT5, STAT3, MAP Kinase, PI3K/AKT

Activates BCL-X

Anti-apoptotic

Cell accumulation

Figure 34-8 Normal regulation of JAK2 function and loss of regulation from the JAK2 V617F mutation. A, Normal function of the JAK2 protein and regula-
tion of phosphorylation by JAK2 intrachain folding and the binding of JAK2 inhibitors SOCS1 and JAB proteins. B, The JAK2 V617F mutation and the loss of
normal JAK2 folding and inhibitor binding resulting in phosphorylation, activation, and the stimulation of STAT, MAP kinase, and PI3K/AKT signal transduction
pathways.

presence (Figure 34-10). Hematopoietic stem cells that bear the low serum erythropoietin levels, and autonomous, in vitro
JAK2 mutation are resistant to erythropoietin-deprivation erythroid colony formation.6 Additional diagnostic features
apoptosis by upregulation of BCL-X, an antiapoptotic protein. of PV include an increased RBC mass of 36mL/kg or greater
PV progenitor cells do not divide more rapidly but accumulate in males and 32mL/kg or greater in females, an arterial
because they do not die normally.55-57 oxygen saturation of 92% (normal) or greater, and spleno-
megaly. Other features of PV are thrombocytosis of greater
Diagnosis than 400 109 platelets/L; leukocytosis of greater than 12
Based on the WHO standards, the diagnosis of PV requires that 109 cells/L without fever or infection; and increases in
two major criteria and one minor criterion be met or that the leukocyte alkaline phosphatase (LAP), serum vitamin B12, or
first major criterion listed and two minor criteria be met. The unbound vitamin B12 binding capacity.58,59 Recent research
two major criteria are an elevated hemoglobin (Hb) level indicates that the JAK2 V617F mutation can be expected in
(greater than 18.5g/dL men and greater than 16.5g/dL in more than 90% to 95% of cases.6 The WHO criteria for the
women) and the identification of the JAK2 V617F mutation, diagnosis of PV are summarized in Box 34-1.
the JAK2 exon 12 mutation, or a similar JAK2 mutation. The It is not always easy to assign an early diagnosis of PV. Eryth-
three minor criteria are panmyelosis in the bone marrow, rocytosis secondary to hypoxia or erythropoietin-producing
CHAPTER 34 Myeloproliferative Neoplasms 519

Unbound Bound EPOR Homo


Inactive Active Dimerization
E O E O E O
P P P E O
P
E O
P
P P
P P E O
P
JAK2 P
1 2 3 JAK2 P
4
P P JAK2 P
Conformational 5 P 6 P
P P
change
STAT3 7 JAK2
JAK2 P P
STAT3 P
8
STAT3

STAT5
Inhibited 13 Increased
apoptosis proliferation
9 STAT3 P STAT5

Protein
Ribosome 12
STAT?

mRNA 11
10 STAT?
Gene Promoter

Nucleus

Figure 34-9 Normal erythropoiesis involving erythropoietin binding to erythropoietin receptors and stimulation of the JAK/STAT pathway via the normal
JAK2 protein.

Unbound Constitutively
Inactive Active JAK2

P P
P P
JAK2 P
1 2 3 JAK2 P
4
P P JAK2 P
Conformational 5 P 6 P P
change P
P STAT3 7 JAK2
P
P P
JAK2
STAT3 P
P 8
STAT3

STAT5
Inhibited 13 Increased
apoptosis proliferation
9 STAT3 P STAT5

Protein
Ribosome 12
STAT?

mRNA 11
10 STAT?
Gene Promoter

Nucleus

Figure 34-10 Stimulation of erythropoiesis in the absence of erythropoietin that is driven by a constitutively phosphorylated and activated JAK2 protein
resulting from the JAK2 V617F mutation.
520 PART V Leukocyte Disorders

TABLE 34-2 Common Morphologic Changes in


BOX 34-1 World Health Organization Criteria for Polycythemia Vera
the Diagnosis of Polycythemia Vera
Diagnosis requires the presence of both major criteria and one minor Peripheral Blood
criterion or the presence of the first major criterion together with two Hemoglobin Increased
minor criteria. Hematocrit Increased
Red blood cell volume Increased
Major Criteria Erythrocyte morphology Normocytic/Normochromic
1. Hemoglobin >18.5g/dL in men, >16.5g/dL in women or other Total white blood cells Increased
evidence of increased red blood cell volume* Granulocytes Increased
2. Presence of JAK2 V617F or other functionally similar mutation such Platelets Increased
as JAK2 exon 12 mutation Leukocyte alkaline phosphatase Normal or increased

Minor Criteria Bone Marrow


1. Bone marrow biopsy specimen showing hypercellularity for age with Normoblasts Increased
trilineage growth (panmyelosis) with prominent erythroid, granulo- Granulocytes Increased
cytic, and megakaryocytic proliferation Megakaryocytes Increased
2. Serum erythropoietin level below the reference range for normal Reticulin Increased
3. Endogenous erythroid colony formation in vitro
Extramedullary Tissue
*Hemoglobin or hematocrit >99th percentile of method-specific reference range
Splenomegaly Present
for age, sex, altitude, or residence, or hemoglobin >17g/dL in men, >15g/dL
in women if associated with a documented and sustained increase of at least Sinusoidal Present
2g/dL from an individuals baseline value that cannot be attributed to correction Medullary Present
of iron deficiency, or elevated red cell mass >25% above mean normal predicted Hepatomegaly Present
value. Sinusoidal Present
From Vardiman JW, Thiele J, Arber DA, et al: The 2008 revision of the World
Health Organization (WHO) classification of myeloid neoplasms and acute leuke-
mia: rationale and important changes. Blood 114:937-951, 2009.

neoplasms is the most difficult to diagnose correctly. In indi-


viduals with these conditions, the bone marrow exhibits ery-
throid hyperplasia without granulocytic or megakaryocytic
hyperplasia. Patients with stress or spurious erythrocytosis
exhibit increased hemoglobin and hematocrit (Hct) without
increased erythrocyte mass or splenomegaly.

Peripheral Blood and Bone Marrow


Common peripheral blood, bone marrow, and tissue findings
in the early or proliferative phase of PV are listed in Table 34-2.
Figures 34-11 and 34-12 show common morphologic patterns
in peripheral blood and bone marrow morphologic and cel- Figure 34-11 Peripheral blood film in stable phase polycythemia vera
lular changes. Not only are quantitative changes seen, but bone with essentially normocytic, normochromic erythrocytes (500).
marrow normoblasts may collect in large clusters, megakaryo-
cytes are enlarged and exhibit lobulated nuclei, and bone
marrow sinuses are enlarged without fibrosis. Pseudo-Gaucher circulatory disturbances increase the risk of hemorrhage, tissue
cells are rare.26 Approximately 80% of patients manifest bone infarction, and thrombosis.
marrow panmyelosis, and 100% of bone marrow volume may
exhibit hematopoietic cellularity. Although the bone marrow Clinical Presentation
pattern may mimic that in other MPNs, the peripheral blood PV initially manifests in a proliferative phase independent of
cells appear normal, with normocytic, normochromic erythro- normal regulatory mechanisms. PV is always associated with
cytes; mature granulocytes; and normal-sized, granulated plate- increased RBC mass. This is the stable phase of PV, which
lets. The other 20% of patients exhibit lesser degrees of progresses to a spent phase in a few patients. In the spent
cellularity in the bone marrow and peripheral blood. Spleno- phase, patients experience progressive splenomegaly (palpable
megaly, hepatomegaly, generalized vascular engorgement, and spleen) or hypersplenism (large spleen with bone marrow
CHAPTER 34 Myeloproliferative Neoplasms 521
Incidence
In the absence of a well-defined diagnostic algorithm, deter-
mining the true incidence of ET has been difficult. When the
diagnostic system developed by the Polycythemia Vera Study
Group (PVSG) is applied, however, the incidence is estimated
to be between 0.6 and 2.5 cases per 100,000 persons per year.
The majority of cases occur in individuals between the ages of
50 and 60 years, but a second peak occurs primarily in women
in the childbearing years, approximately 30 years of age.63

Clinical Presentation
In more than one half of the patients diagnosed with ET, the
disorder is discovered in the laboratory by virtue of an unex-
pectedly elevated platelet count on a routine complete blood
Figure 34-12 Bone marrow biopsy specimen in stable phase polycythe- count; the remaining patients see a physician due to vascular
mia vera showing panmyelosis (hematoxylin and eosin stain, 400). occlusion or hemorrhage. Vascular occlusions are often the
result of microvascular thromboses in the digits or thromboses
in major arteries and veins that occur in a variety of organ
hyperplasia and peripheral blood cytopenias) and pancytope- systems, including splenic or hepatic veins as in Budd-Chiari
nia. They may also exhibit the triad of bone marrow fibrosis, syndrome. Bleeding occurs most frequently from mucous
splenomegaly, and anemia with teardrop-shaped poikilocytes. membranes in the gastrointestinal and upper respiratory tracts.
The latter pattern is called postpolycythemic myeloid metaplasia, Splenomegaly is observed at presentation in 50% of patients
and its morphologic features are similar to those of PMF. when the PVSG diagnostic criteria are used but at a much
Peripheral WBC and RBC counts vary, and nucleated erythro- lower frequency when the WHO standards are applied. This
cytes, immature granulocytes, and large platelets are present. difference is largely due to the elimination of the diagnosis
Usually, splenomegaly is secondary to extramedullary hemato- of ET in patients who meet the criteria for PMF in the prefi-
poiesis.60 Myelofibrosis occurs within the bone marrow and brotic stage.63
may come to occupy a significant proportion of bone marrow
volume, with subsequent ineffective hematopoiesis.61 Diagnosis
ET must be differentiated from secondary or reactive thrombo-
Treatment and Prognosis cytoses and from other MPNs. Thrombocytosis may be second-
The disease progresses to acute leukemia in 15% of patients. ary to chronic active blood loss, hemolytic anemia, chronic
The use of myelosuppressive therapy such as phosphorus P 32 inflammation or infection, or nonhematogenous neoplasia.
(32P) or alkylating agents seems to increase the risk.59 Only 1% The diagnostic criteria for ET first proposed by the PVSG were
to 2% of patients treated with phlebotomy alone experience intended to distinguish ET from other MPNs. These features
leukemic transformation. However, the risk of thrombosis and included a platelet count of greater than 600 109/L, a hemo-
bleeding is increased in patients treated with phlebotomy globin of less than 13g/dL or a normal erythrocyte mass, and
alone, so the use of alkylating myelosuppressive agents may be stainable iron in the bone marrow or a failure of iron therapy.
required to control these complications. Some patients may Philadelphia chromosome negativity, absence of marrow
manifest a temporary disease pattern similar to myelodyspla- collagen fibrosis (less than one third of a biopsy specimen
sia, and the cell morphology in transformation to acute leuke- is fibrous), no splenomegaly, absence of leukoerythroblastic
mia may be difficult to classify. Patients with both early and reaction, and no known cause of reactive thrombocytosis all
advanced PV may show clinical, peripheral blood, bone support the diagnosis.64
marrow, and extramedullary features that mimic those of The newest WHO group now requires the documentation
other MPNs. of four major criteria to establish a diagnosis of ET. First, the
WHO group lowered the platelet count threshold to 450
109/L or greater to capture patients who would eventually meet
ESSENTIAL THROMBOCYTHEMIA
diagnostic criteria but who were experiencing hemorrhage or
Essential thrombocythemia (ET) is a clonal MPN with thrombosis with platelet counts of between 450 109/L and
increased megakaryopoiesis and thrombocytosis, usually 600 109/L.63 Because a lower platelet threshold could lead to
with a count greater than 600 109/L and sometimes with a false-positive ET diagnoses, all the WHO criteria must be met
count greater than 1000 109/L.62 However, WHO criteria to eliminate such patients. Second, the bone marrow must
require a sustained thrombocytosis with a platelet count of show significant megakaryopoiesis characterized by large,
450 109/L or greater. Over the years, ET has been known as mature-looking megakaryocytes with no substantial increase in
primary thrombocytosis, idiopathic thrombocytosis, and hemorrhagic erythropoiesis or granulopoiesis or left shift in the neutrophil
thrombocythaemia.63 line. Third, the condition cannot meet the criteria of any other
522 PART V Leukocyte Disorders

MPN, myelodysplasia, or other myeloid neoplasm. Fourth, density (Figure 34-14). Special studies reveal increased numbers
patients must demonstrate either the JAK2 V617F or other of smaller and less mature megakaryocytes.68 Increased granu-
clonal mutation or, in the absence of a clonal marker, the lopoiesis and erythropoiesis may contribute to bone marrow
absence of reactive thrombocytosis. Careful analysis of the hypercellularity, and in a few patients, reticulin fibers may be
bone marrow biopsy specimen is useful in distinguishing ET increased. The major peripheral blood, bone marrow, and
from myelodysplastic syndromes (MDSs) associated with the extramedullary findings are listed in Table 34-3.
del(5q) mutation, refractory anemia with ringed sideroblasts
with thrombocytosis, and the prefibrotic phase of PMF. Like-
wise, the identification of the JAK2 V617F mutation excludes
cases of reactive thrombocytosis.63 TABLE 34-3 Common Morphologic Changes in
The JAK2 V617F mutation is found in 50% to 60% of ET Essential Thrombocythemia
patients and supports the diagnosis of ET.65,66 WHO diagnostic Peripheral Blood
criteria were modified to include a minimum platelet count Hemoglobin Slightly decreased
of 450 109/L when the JAK2 mutation is present.67 JAK2 Hematocrit Slightly decreased
mutations have not been identified in the germline of any Red blood cell volume Normal
patient with MPN disorder, which supports the view that the Total white blood cells Normal or slightly increased
mutation is acquired. MPL W515K/L, a mutation in the throm- Neutrophils Normal or slightly increased
bopoietin receptor (MPL), has been reported in 1% of ET Platelets Increased
cases and is also used to exclude a diagnosis of reactive throm- Platelet function Decreased
bocytosis. Other genetic mutations are uncommon but have
been reported to be found in 5% to 10% of cases when the Bone Marrow
diagnostic criteria proposed by the PVSG are applied. The Normoblasts Normal or increased
most commonly reported additional mutations are +8, 9q, and Granulocytes Normal or slightly increased
(del)20q.63 Megakaryocytes
Clusters Present
Peripheral Blood and Bone Marrow Large Present
Figure 34-13 shows a peripheral blood film that exhibits early- Hyperlobulated Present
phase thrombocytosis with variation in platelet diameter and Dense nuclei Present
shape, including giantism, agranularity, and pseudopods. Variability in size Increased
Commonly, platelets are present in clusters and tend to accu- Reticulin Normal or slightly increased
mulate near the thin edge of the blood film. Segmented neu-
trophils may be increased; basophils are not. Erythrocytes are Extramedullary Tissue
normocytic and normochromic, unless iron deficiency is Splenomegaly Present
present secondary to excessive bleeding. Sinusoidal Present
Early-phase bone marrow shows marked megakaryocytic Medullary Present
hypercellularity, clustering of megakaryocytes, and increased Megakaryocytic proliferation Present
megakaryocyte diameter with nuclear hyperlobulation and

Figure 34-13 Peripheral blood film in stable phase essential thrombocy- Figure 34-14 Bone marrow biopsy specimen in essential thrombocythe-
themia showing increased numbers of platelets and mature neutrophils mia showing marked megakaryocytic hypercellularity (hematoxylin and eosin
(500). stain, 400).
CHAPTER 34 Myeloproliferative Neoplasms 523

A diagnosis of ET is questionable in patients with a platelet normoblasts, dacryocytes (teardrop-shaped RBCs), and other
count of more than 450 109/L if certain features are observed bizarre RBC shapes.
on the bone marrow biopsy specimen. For example, increased PMF clonality was manifest in studies in which cytogenetic
erythropoiesis or granulopoiesis in the bone marrow is a ques- abnormalities were detected in normoblasts, neutrophils,
tionable finding for ET and suggests an alternative diagnosis of macrophages, basophils, and megakaryocytes. Female patients
PV or PMF, respectively, especially if bizarre or significantly heterozygous for glucose-6-phosphate dehydrogenase isoen-
atypical megakaryocytes are also observed. Dyserythropoiesis zymes have PMF cells of a single enzyme isotype, whereas
and/or dysgranulopoiesis suggests a myelodysplastic disorder tissue cells, including marrow fibroblasts, contain both enzyme
and should prompt an investigation for (del)5q, (inv)3, and/ isotypes.4
or t(3;3).63

Myelofibrosis
Treatment and Prognosis The myelofibrosis in this disease comprises three of the five
Patients with ET experience relatively long survival provided types of collagen: I, III, and IV. Increases in type III collagen
they remain free of serious thromboembolic or hemorrhagic are detected by silver impregnation techniques, increases in
complications. Clinical symptoms associated with throm type I by staining with trichrome, and increases in type IV by
boembolic vasoocclusive events include the syndrome of the presence of osteosclerosis, which may be diagnosed from
erythromelalgia (throbbing and burning pain in the hands and increased radiographic bone density.69 In approximately 30%
feet, accompanied by mottled redness of areas), transient isch- of patients, biopsy specimens show no fibrosis.37 Increases in
emic attacks, seizures, and cerebral or myocardial infarction. these collagens are not a part of the clonal proliferative process
Other symptoms include headache, dizziness, visual distur- but are considered secondary to an increased release of fibro-
bances, and dysesthesias (decreased sensations). Hemorrhagic blastic growth factors, such as platelet-derived growth factor,
complications include bleeding from oral and nasal mucous transforming growth factor from megakaryocyte -granules,
membranes or gastrointestinal mucosa and the appearance of tumor necrosis factor-, and interleukin-1, and interleukin-
cutaneous ecchymoses (see Chapter 43). 1. Marrow fibrosis causes expansion of marrow sinuses and
Treatment involves prevention or early alleviation of hem- vascular volume with an increased rate of blood flow. Bone
orrhagic or vasoocclusive complications that occur as the plate- marrow fibrosis is not the sole criterion for the diagnosis of
let count increases. The production of platelets must be reduced PMF, because increases in marrow fibrosis may reflect a repara-
by suppressing marrow megakaryocyte production with 32P or tive response to injury from benzene or ionizing radiation,
one of several alkylating agents. As observed in PV, ET patients may be a consequence of immunologically mediated injury,
so treated may incur an increased risk for disease transforma- or may represent a reactive response to other hematologic
tion to acute leukemia or myelofibrosis. However, malignant conditions.
transformation occurs at a frequency of less than 5%.63 Type IV collagen and laminin normally are discontinuous
Hydroxyurea therapy may achieve a desired reduction of in sinusoidal membranes but appear as stromal sheets in
peripheral platelets without the risk of complications experi- association with neovascularization and endothelial cell pro-
enced with myelosuppressive agents. This may relate to the liferation in regions of fibrosis. In addition, deposition of
youth of ET patients, in whom the risk of leukemic transforma- type VII collagen is observed, and this may form a linkage
tion seems relatively low. More recently, cytoreduction has between type I fibers, type III fibers, and type I plus type III
been achieved with interferon-. fibers.70
Studies commonly show a median survival of longer
than 10 years, including cases in which the process arises
in younger patients. However, some patients may develop Hematopoiesis and
post-ET myelofibrosis, which reduces survival. Patients whose Extramedullary Hematopoiesis
cells manifest chromosome abnormalities may have a poorer Extramedullary hematopoiesis, clinically recognized as hepato-
prognosis.37 megaly or splenomegaly, seems to originate from release of
clonal stem cells into the circulation.71 The cells accumulate in
the spleen, liver, or other organs, including adrenals, kidneys,
lymph nodes, bowel, breasts, lungs, mediastinum, mesentery,
PRIMARY MYELOFIBROSIS
skin, synovium, thymus, and lower urinary tract. The cause of
Primary myelofibrosis (PMF), previously known as chronic extramedullary hematopoiesis is unknown. In experimental
idiopathic myelofibrosis, agnogenic myelofibrosis, and myelofibrosis animal models, chemicals, hormones, viruses, radiation, and
with myeloid metaplasia, is a clonal MPN4 in which there is immunologic factors have been implicated. The disease is asso-
splenomegaly and ineffective hematopoiesis associated with ciated with an increase in circulating hematopoietic cells, but
areas of marrow hypercellularity, fibrosis, and increased mega- fibroblasts are a secondary abnormality and not clonal.72 B and
karyocytes. Megakaryocytes are enlarged with pleomorphic T cells may be involved.73 There is an increase in circulating
nuclei, coarse segmentation, and areas of hypochromia. The unilineage and multilineage hematopoietic progenitor cells,74
peripheral blood film exhibits immature granulocytes and and the number of CD34+ cells may be 300 times normal.75
524 PART V Leukocyte Disorders

The increase in circulating CD34+ cells separates PMF from TABLE 34-4 Common Morphologic Changes in Primary
other MPNs and predicts the degree of splenic involvement Myelofibrosis
and risk of conversion to acute leukemia.
Body cavity effusions containing hematopoietic cells may
Peripheral Blood
Hemoglobin Normal or decreased
arise from extramedullary hematopoiesis in the cranium, the
Anisocytosis Present
intraspinal epidural space, or the serosal surfaces of pleura,
Poikilocytosis Present
pericardium, and peritoneum. Portal hypertension, with its
Teardrop-shaped erythrocytes Present
attendant consequences of ascites, esophageal and gastric
Nucleated red blood cells Present
varices, gastrointestinal hemorrhage, and hepatic encephalopa-
Polychromasia Normal or increased
thy, arises from the combination of a massive increase in
Total white blood cells Normal, decreased, or increased
splenoportal blood flow and a decrease in hepatic vascular
Immature granulocytes Increased
compliance secondary to fibrosis around the sinusoids and
Blasts Present
hematopoietic cells within the sinusoids.76
Basophils Present
Leukocyte anomaly Present
Incidence and Clinical Presentation
Leukocyte alkaline phosphatase Increased, normal, or decreased
The disease occurs in patients older than age 60 and may
Platelets Increased, normal, or decreased
be asymptomatic. PMF generally presents with fatigue, weak-
Abnormal platelets Present
ness, shortness of breath, palpitations, weight loss, and dis-
Megakaryocytes Present
comfort or pain in the left upper quadrant associated with
splenomegaly.
Bone Marrow
Cellularity Increased
Peripheral Blood and Bone Marrow
Granulopoiesis Increased
PMF presents with a broad range of changes in laboratory test
Megakaryocytes Increased
values and peripheral blood film results, but examination of
Erythropoiesis Normal or increased
the bone marrow biopsy specimen provides most of the infor-
Myelofibrosis Increased
mation for diagnosis. Changes commonly observed in periph-
Sinuses Increased
eral blood and bone marrow examinations are summarized in
Dysmegakaryopoiesis Present
Table 34-4.
Dysgranulopoiesis Present
Abnormalities in erythrocytes noted on peripheral blood
films include the presence of dacryocytes, other bizarre shapes,
nucleated RBCs, and polychromatophilia. Granulocytes are
Extramedullary Tissue
Splenomegaly Present
increased, normal, or decreased in number and may include
Sinusoidal Present
immature granulocytes, blasts, and cells with nuclear or cyto-
Medullary Present
plasmic anomalies. Platelets may be normal, increased, or
Hepatomegaly Present
decreased in number with a mixture of normal and abnormal
Sinusoidal Present
morphologic features (Figure 34-15). Micromegakaryocytes
Portal tract Present
may be observed (Figure 34-16).
Local infiltrates Present
Bone marrow biopsy specimens exhibit intense fibrosis,
Other tissues Present
granulocytic and megakaryocytic hypercellularity, dysmega-
karyopoiesis, dysgranulopoiesis, and numerous dilated sinuses
containing luminal hematopoiesis. Neutrophils may exhibit
impairment of physiologic functions such as phagocytosis,
oxygen consumption, and hydrogen peroxide generation,
and decreased myeloperoxidase and glutathione reductase
activities. Platelets show impaired aggregation in response
to epinephrine, decreased adenosine diphosphate concentra-
tion in dense granules, and decreased activity of platelet
lipoxygenase.

Immune Response
Humoral immune responses are altered in approximately 50%
of patients and include the appearance of autoantibodies to
erythrocyte antigens, nuclear proteins, gamma globulins, phos-
pholipids, and organ-specific antigens.77 Circulating immune
complexes, increased proportions of marrow-reactive lympho-
cytes, and the development of amyloidosis are evidence for Figure 34-15 Peripheral blood film in primary myelofibrosis showing
active immune processes. Collagen disorders coexist with PMF, nucleated red blood cells, giant platelets, and immature myeloid cells (1000).
CHAPTER 34 Myeloproliferative Neoplasms 525

ment to date for patients younger than age 60 is allogeneic


stem cell transplantation. Five-year survival approaches 50%
in patients undergoing transplantation, but 1-year mortality is
27%, and graft-versus-host disease occurs in 33%.80

INTERCONNECTION BETWEEN ESSENTIAL


THROMBOCYTHEMIA, POLYCYTHEMIA
VERA, AND PRIMARY MYELOFIBROSIS
The discovery of the JAK2 V617F mutation has advanced our
understanding of the MPNs but has also raised questions about
the interconnection of three of the primary myeloproliferative
conditions: ET, PV, and PMF. Why is the JAK2 mutation found
Figure 34-16 Peripheral blood film in primary myelofibrosis exhibiting in more than 90% to 95% of patients with PV but in only 50%
increased platelets and a micromegakaryocyte (1000). to 60% of patients with ET and PMF, and how can the same
mutation produce three distinct phenotypes?
Currently four hypotheses exist to account for this apparent
discordance. One prevailing thought suggests that the resulting
which suggests that immunologic processes may stimulate phenotype is dependent on the stage of differentiation of the
marrow fibroblast activity. hematopoietic stem cell. For example, if the hematopoietic
stem cell has developed a predilection toward platelet develop-
ment at the time of the JAK2 mutation, ET will develop. Reports
Treatment and Prognosis have described differences in differentiation programs81,82 and
Average survival from the time of diagnosis is about 5 years, in JAK2 mutations among ET, PV, and PMF.83 A second hypoth-
but patients have lived 15 years. During this time, increasing esis proposes that the genetic background of the patient pre-
numbers and pleomorphy of megakaryocytes lead to progres- disposes the patient to a particular phenotype. Mutations in
sive marrow failure. Marrow blasts may increase.37 Adverse the erythropoietin receptor, thrombopoietin receptor (MPL),
prognostic indicators include more severe anemia and throm- and granulocyte colony-stimulating factor receptor have all
bocytopenia, greater hepatomegaly, unexplained fever, and been implicated.84 The third hypothesis suggests that the phe-
hemolysis. Mortality is associated with infection, hemorrhage, notype depends of the level of JAK2 tyrosine kinase activity,
postsplenectomy complications, and transformation to acute called the dosage effect. Patients diagnosed with PV showed
leukemia. greater tyrosine kinase activity than patients presenting with an
A diverse spectrum of therapies has been implemented to ET phenotype. Experiments in which erythroid progenitors
alleviate symptoms or modify clinical problems in these were collected from these patients and tested in colony-forming
patients. Severe anemia has been treated with androgen therapy, assays showed that nearly all the cell cultures developing a PV
and hemolytic anemia with glucocorticosteroids. Reduction phenotype were homozygous for the JAK2 mutation, whereas
of myelofibrosis and of marrow and tissue hypercellularity the ET phenotype was observed in the vast majority of
has been accomplished with busulfan and other alkylating cell cultures expressing a heterozygous genotype.85,86 This phe-
agents, hydroxyurea, and, in a few patients, interferon- and nomenon was corroborated in experiments with transgenic
interferon-. Radiotherapy is considered for patients with severe mice.87,88 The last hypothesis proposes that a pre-JAK2 muta-
splenic pain, for patients with massive splenomegaly who are tion produces a premalignant clone89-91 predisposing the hema-
not clinical candidates for splenectomy, for patients with ascites topoietic stem cell to a particular phenotype and that the JAK2
secondary to serosal implants (metastatic nodules), and for mutation drives the malignant transformation. Groups have
patients with localized bone pain and localized extramedullary reported mutations coexisting with the JAK2 V617F mutation,
fibrohematopoietic masses in other areas, especially in the like BCR/ABL,92,93 MPL mutations,94 and another version of
epidural space. Splenectomy is performed to end severe pain, the JAK2 mutation.95,96 Familial MPN provides the strongest
excessive transfusion requirements, or severe thrombocytope- support for a pre-JAK2 mutation.97,98
nia and to correct severe portal hypertension. The most appealing model includes all of the hypotheses
Chemotherapy is partially successful in reducing the number previously presented and suggests that ET, PV, and PMF may
of CD34+ cells and immature hematopoietic cells, marrow fibro- represent a continuum of diseases. It seems reasonable to
sis, and splenomegaly.78 Single-agent chemotherapy is most assume that a pre-JAK2 mutation occurs in the hematopoietic
helpful in the early clinical phases of the disease, and agents stem cell most if not all of the time to create a hyperprolifera-
such as busulfan, 6-thioguanine, and chlorambucil, alone or in tive clone that predisposes to additional mutations like the
combination with other chemotherapy, are useful. Other thera- JAK2 mutation. The pre-JAK2 mutation can be familial, con-
pies include interferon-, hydroxyurea, and combinations of genital, or somatic. Because MPL is expressed in high levels
the previously mentioned drugs.79 The most successful treat- on megakaryocyte precursors, one JAK2 V617F mutation
526 PART V Leukocyte Disorders

(heterozygous) is sufficient to induce MPL signaling and thus blood. The bone marrow shows an increase in normal-
stimulate megakaryocyte production. This could lead to the ET appearing neutrophilic cells with fewer than 5% myeloblasts.
phenotype. In contrast, the erythropoietin receptor is expressed Megakaryocytes are normal or slightly left-shifted. Splenomeg-
in low density on the surface of erythroid precursors, which aly must be present and is often accompanied by hepatomeg-
requires the higher amount of JAK2 V617F tyrosine kinase that aly. Reactive neutrophilia must be excluded by eliminating
is produced by two JAK2 mutations (homozygous). This could infection, inflammation, and tumors as a cause of the neutro-
lead to the PV phenotype. Because MPL stimulation begins philia. A diagnosis of CNL can still be made in the presence of
with the first JAK2 mutation and continues with the second a reactive process if clonality of the myeloid line can be docu-
JAK2 mutation, the MPL receptor undergoes continuous stimu- mented by karyotyping or molecular analysis. There must be
lation. It has been shown that excessive thrombopoietin stimu- no evidence of the Philadelphia chromosome, BCR/ABL muta-
lation leads to myelofibrosis, which may result in a progression tion, or rearrangements of the PDGFRA, PDGFRB, or FGRF1
to PMF.99 genes. Lastly, there can be no evidence of PV, PMF, ET, MDS,
or MDS/MPN disorders.100

OTHER MYELOPROLIFERATIVE NEOPLASMS


Genetics
Chronic Neutrophilic Leukemia Approximately 90% of CNL patients have a normal karyotype,
Chronic neutrophilic leukemia (CNL) is a clonal disorder in but chromosomal abnormalities are observed, particularly as
which a hyperproliferation of neutrophilic cells in the bone the disease progresses, including +8, +9, +21, del(20q),
marrow produces sustained neutrophilia in the peripheral del(11q), and del(12p). The Philadelphia chromosome or a
blood and hepatosplenomegaly. CNL must be differentiated BCR/ABL mutation cannot be expressed in CNL; otherwise a
from CML, based on the absence of the Philadelphia chromo- diagnosis of CML is required. JAK2 mutations have been
some and the BCR/ABL fusion gene, as well as from both a observed but rarely.100
reactive neutrophilic process and other MPNs.100
Prognosis
Incidence CNL is a slow, smoldering condition, and patient survival
The incidence of CNL is not known, but it is a rare disorder of ranges from as short as 6 months to longer than 20 years. The
which about 150 cases have been reported. However, if the neutrophilia does progress, and some patients develop myelo-
WHO criteria had been applied in these cases, many might dysplasia and can experience transformation into AML.100
have been reclassified as reactive rather than neoplastic
conditions.100 Chronic Eosinophilic Leukemia,
Not Otherwise Specified
Clinical Presentation Chronic eosinophilic leukemia (CEL) is a clonal proliferation
Hepatosplenomegaly is the most common finding, but 25% to of eosinophils from eosinophil precursors that dominate in the
30% of patients report bleeding from mucocutaneous sites like bone marrow and peripheral blood. Eosinophils are found in
the gastrointestinal tract. Other symptoms include gout from other peripheral tissues, including heart, lungs, central nervous
WBC turnover and pruritus that may be associated with neu- system, gastrointestinal tract, and skin. Hepatosplenomegaly is
trophil infiltration of tissues and organs.100 observed in approximately 30% to 50% of patients. Infiltrating
eosinophils degranulate to release cytokines, enzymes, and
Peripheral Blood and Bone Marrow other granular proteins that damage the surrounding tissue,
Patients have a WBC count of more than 25 109/L with a which results in organ dysfunction.101
slight left shift. Neutrophils dominate but the increase in
bands, metamyelocytes, myelocytes, and promyelocytes in Clinical Presentation
combination usually comprise fewer than 5% of WBCs but can Although some patients may be asymptomatic when found to
be as many as 10%. Neutrophils do not appear dysplastic, but have eosinophilia, most have signs and symptoms of fever,
they often contain toxic granules. RBC and platelet morphol- fatigue, cough, angioedema, muscle pain, and pruritus. A more
ogy are normal in the peripheral blood.100 severe sequela of CEL involves the heart. Fibrosis can form in
The bone marrow reflects the peripheral blood in that it is the heart (endomyocardial fibrosis), which can evolve into
hypercellular with predominately a proliferation of neutro- cardiomegaly. Within the heart scar tissue may form in the
phils, including myelocytes, metamyelocytes, bands, and seg- mitral and tricuspid valves, affecting valve function and predis-
mented neutrophils. The myeloid-to-erythroid ratio is at least posing to thrombi formation. Other serious complications
20:1 or greater. RBCs and platelets are normal in number, and include peripheral neuropathy, central nervous system dysfunc-
no cell line exhibits significant dysplastic morphology.100 tion, pulmonary symptoms from eosinophilic infiltrates, and
rheumatologic problems.101
Diagnosis
The WBC count must be greater than 25 109/L with greater Peripheral Blood and Bone Marrow
than 90% mature neutrophils, fewer than 10% immature Peripheral eosinophilia must be observed, with the majority of
neutrophilic cells, and fewer than 1% blasts in the peripheral eosinophils appearing normal. Some evidence of eosinophil
CHAPTER 34 Myeloproliferative Neoplasms 527
abnormality is found, however, and includes the presence of before 6 months of age. In contrast, systemic mastocytosis
eosinophilic myelocytes and metamyelocytes, hypogranula- generally occurs after the second decade of life. Approximately
tion, and vacuolization. Neutrophilia is a common finding; 80% of patients with mastocytosis show skin involvement
other features such as mild monocytosis, basophilia, and the regardless of the type of mastocytosis diagnosed. Cutaneous
presence of blasts are less common. The bone marrow is hyper- mastocytosis occurs in the skin; systemic mastocytosis usually
cellular owing to eosinophilic proliferation and can demon- involves the bone marrow and other organ systems like the
strate Charcot-Leyden crystals. Myeloblast numbers are elevated spleen, lymph nodes, liver, and gastrointestinal tract; and mast
but below the 20% threshold necessary to classify the disorder cell leukemia is characterized by mast cells in the peripheral
as an acute leukemia. Erythrocytes and megakaryocytes are blood.102
normal in number but sometimes demonstrate dysplastic mor-
phologic features. Bone marrow fibrosis occurs due to the Clinical Presentation
release of eosinophilic basic protein and eosinophilic cationic Patients present with urticarial lesions (wheel and flare) that
proteins from the eosinophil granules. Bone marrow fibrosis may become activated when stroked upon physical examina-
contributes to the premature release of eosinophils into the tion. Skin lesions also tend to have melanin pigmentation.
circulation, and they deposit in a variety of tissues.101 Four categories of symptom severity have been described in
mastocytosis: (1) constitutional systems like fatigue and weight
Diagnosis loss; (2) skin manifestations; (3) mediator-related systemic
The diagnosis of CEL requires eosinophilia with a count of events such as abdominal pain, gastrointestinal distress, head-
more than 1.5 109 cells/L and the presence of malignant ache, and respiratory symptoms; and (4) musculoskeletal
features, and the elimination of reactive eosinophilia and other complaints like bone pain, arthralgias, and myalgias. Hemato-
malignancies that have concomitant eosinophilia. Reactive logic findings include anemia, leukocytosis, eosinophilia, neu-
conditions like parasitic infections, allergies, Loeffler syndrome tropenia, and thrombocytopenia. In patients with systemic
(pulmonary disease), cyclical eosinophilia, angiolymphoid mastocytosis with associated clonal hematologic nonmast-
hyperplasia of the skin, collagen vascular disorders, and Kimura cell-lineage disease the most common associated hematologic
disease must be excluded in the differential diagnosis. Likewise, finding is chronic myelomonocytic leukemia, but any myeloid
other malignancies that can produce a concomitant eosino- or lymphoid malignancy can occur, although myeloid versions
philia include T-cell lymphoma, Hodgkin lymphoma, systemic predominate.102
mastocytosis, chronic myelomonocytic leukemia, atypical
CML, and ALL. These disorders lead to the release of a variety Diagnosis
of interleukins that can drive a secondary eosinophil reaction. The typical skin lesion is the first diagnostic clue to mastocy-
No single genetic abnormality is specific for CEL, but CEL can tosis. Cutaneous mastocytosis occurs in three forms: urticaria
be ruled out by the presence of several karyotypic abnormali- pigmentosa, diffuse cutaneous mastocytosis, and mastocytosis
ties, such as PDGFRA, PDGFRB, FGFR1, and BCR/ABL. of the skin, all of which occur predominantly in children. In
Common myeloid mutations like +8, i(17q), and JAK2 can urticaria pigmentosa mast cells are confined to the skin and
support a diagnosis of CEL.101 form aggregates in the dermis, whereas in diffuse cutaneous
mastocytosis, mast cells are found in more than one cutaneous
Prognosis location. In systemic mastocytosis, mast cells are observed in
Survival is variable, but approximately 80% of patients will live one area of the bone marrow with fibrosis and other areas of
5 years after diagnosis. Features of dysplasia, an increase in the bone marrow are hypercellular with panmyelosis, and/or
karyotype abnormalities, or an increase in blasts indicates an mast cells are identified in other extracutaneous sites. In addi-
unfavorable prognosis.101 tion to the major criteria just described, at least one of four
minor criteria must be met. These include the following:
Mastocytosis (1) More than 25% of mast cells must be immature or have
Mastocytosis is a broad term referring to a clonal neoplastic atypical morphology like a spindle shape. (2) Mast cells must
proliferation of mast cells, which accumulate in one or more express a KIT mutation at codon 816. (3) Mast cells must
organ systems, but it can present differently and manifest in a express normal markers and CD2 and/or CD25. (4) Total
range of severities. The WHO group has classified mastocytosis serum tryptase must be above 20mg/mL. The key diagnostic
into seven subcategories: cutaneous mastocytosis, indolent features that separate the six types of systemic mastocytosis
systemic mastocytosis, systemic mastocytosis with associated are as follows: (1) Indolent systemic mastocytosis is character-
clonal hematologic nonmast-cell-lineage disease, aggressive ized by a low mast cell burden. (2) Systemic mastocytosis
systemic mastocytosis, mast cell leukemia, mast cell sarcoma, with associated clonal hematologic nonmast-cell-lineage
and extracutaneous mastocytoma.102 disease presents with one of the following WHO classifications
of neoplasms: myelodysplastic syndrome, myeloproliferative
Incidence neoplasm, AML, lymphoma, or other hematopoietic neo-
Mastocytosis can occur at any age, with cutaneous mastocytosis plasm. (3) Aggressive systemic mastocytosis usually does not
seen most often in children. Babies can be born with cutaneous manifest with skin lesions or mast cells in circulation but does
mastocytosis, and half of affected children develop the disease have one or more of the following: mast cells in bone marrow,
528 PART V Leukocyte Disorders

dysplastic hematopoietic changes, or hepatosplenomegaly. but may survive only a few months after diagnosis. Signs and
(4) Mast cell leukemia is characterized by more than 20% symptoms that predict a poorer prognosis include elevated
atypical mast cells in the bone marrow and more than 10% lactate dehydrogenase and alkaline phosphatase, anemia,
in the peripheral blood. (5) Mast cell sarcoma presents as a thrombocythemia, abnormal peripheral blood morphology,
single unifocal mast cell tumor with a high-grade pathology. bone marrow hypercellularity, and hepatosplenomegaly.102
(6) Extracutaneous mastocytoma also exhibits a unifocal mast
cell tumor, but it is of low-grade pathology.102 Myeloproliferative Neoplasm, Unclassifiable
The category myeloproliferative neoplasm, unclassifiable (MPN-U)
Genetics is designed to capture disorders that clearly express myelopro-
The most common genetic mutation in patients with mastocy- liferative features but either fail to meet the criteria of a specific
tosis involves codon 816 in the KIT gene and occurs in about condition or have features that overlap two or more specific
95% of adults and 33% of children with systemic mastocytosis. conditions. Most patients with MPN-U fall into one of three
This mutation replaces aspartic acid with valine, which alters groups: (1) patients with an early stage of PV, ET, or PMF in
the tyrosine kinase receptor activity so as to cause constitutive which the criteria that define the disorders are not yet fully
kinase activity in the absence of ligand. Usually the mutation developed; (2) patients presenting with features indicative of
is somatic, but a few cases of familial KIT mutations have been advanced disease resulting from clonal evolution that masks
reported. Additional mutations can push proliferation of the potential underlying condition; and (3) patients who have
hematopoietic clones, causing systemic mastocytosis with clear evidence of an MPN but who have a concomitant condi-
associated clonal hematologic nonmast cell lineage disease. tion like a second neoplasm or an inflammatory condition that
These include mutations of RUNX1/RUNX1T in AML, JAK1 alters the MPN features. MPN-U may account for as many as
in MPN, and FIP1L1/PDGFRA in myeloid neoplasms with 10% to 15% of MPN disorders, but caution should be exercised
eosinophilia.102 so that morphologic changes caused by the patients cytotoxic
drug therapy or growth factor therapy, or poor collection of
Prognosis samples are not confused with the features of MPN-U. In
Cutaneous mastocytosis in children has a favorable prognosis patients with MPN-U in the early stages of development or
and may regress spontaneously around puberty. Milder ver- with a concomitant disorder like inflammation, the MPN-U
sions like cutaneous mastocytosis and indolent cutaneous may be reclassified to a specific category of MPN once the
mastocytosis follow a benign course and are associated with a disease begins to express typical features or the secondary
normal life span. Hematologic involvement usually evolves conditions subsides. Likewise, in patients with an advanced
into the corresponding hematologic disease. Patients with MPN, the disorder may be reclassified as an acute leukemia
aggressive systemic mastocytosis, mast cell leukemia, and mast once the blast criterion of more than 20% blasts in the bone
cell sarcoma are often treated with cytoreductive chemotherapy marrow is met.103

SUMMARY
MPNs are clonal hematopoietic stem cell disorders that result in Bone marrow transplantation has been successful in CML, and
excessive production and overaccumulation of erythrocytes, gran- imatinib mesylate, a tyrosine kinase inhibitor, produces remission
ulocytes, and platelets in some combination in bone marrow, in most cases.
peripheral blood, and body tissues. Approximately 4% of CML patients given imatinib as first-line
Within the classification of MPN the four major conditions are CML, therapy develop imatinib resistance.
PV, ET, and PMF. Dosage escalation or administration of second-generation tyrosine
In CML, there are large numbers of myeloid precursors in the bone kinase inhibitors restores remission in most patients with imatinib
marrow, peripheral blood, and extramedullary tissues. resistance.
The peripheral blood exhibits leukocytosis with increased myeloid PV manifests with panmyelosis in the bone marrow with increases
series, particularly the later maturation stages, often with increases in erythrocytes, granulocytes, and platelets.
in eosinophils and basophils. The clinical diagnosis of PV requires a hemoglobin of greater than
The LAP score is dramatically decreased in CML. 18.5g/dL in men and greater than 16.5g/dL in women or other
The Philadelphia chromosome, t(9;22), either at the chromosomal evidence of increased RBC volume and the presence of JAK2
or molecular level, must be present in all cases of CML. V617F or another mutation in the JAK2 gene. If only one of these
In CML, the bone marrow exhibits intense hypercellularity with a major criteria is met, two of the minor criteria must be satisfied.
predominance of myeloid precursors. Megakaryocyte numbers are The JAK2 V617F mutation is found in 90% to 95% of PV patients
normal to increased. and contributes to the pathogenesis of the disease.
Patients with CML progress from a chronic stable phase through ET involves an increase in megakaryocytes with a sustained plate-
an accelerated phase into transformation to acute leukemia. let count greater than 450 109/L.
CHAPTER 34 Myeloproliferative Neoplasms 529

Other diagnostic criteria include normal RBC mass, stainable iron The peripheral blood in PMF exhibits immature granulocytes
in the bone marrow, absence of the Philadelphia chromosome, and nucleated RBCs; teardrop-shaped cells are a common
lack of marrow collagen fibrosis, absence of splenomegaly or finding.
leukoerythroblastic reaction, and absence of any known cause of Platelets may be normal, increased, or decreased in number with
reactive thrombocytosis. abnormal morphology. Micromegakaryocytes may be present.
In the early phases of ET, peripheral blood shows increased Immune responses are altered in about 50% of patients.
numbers of platelets with abnormalities in size and shape. Bone Other MPNs include CNL, CEL not otherwise specified, mastocy-
marrow megakaryocytes are increased in number and in size. tosis, and unclassifiable MPN.
Complications of ET include thromboembolism and hemorrhage.
The JAK2 V617F mutation is observed in 50% to 60% of patients Now that you have completed this chapter, go back and
with ET and PMF and contributes to the pathogenesis of the read again the case study at the beginning and respond
disorders. to the questions presented.
PMF manifests with ineffective hematopoiesis, sparse areas of
marrow hypercellularity (especially with increased megakaryo-
cytes), bone marrow fibrosis, splenomegaly, and hepatomegaly.

R E V I E W Q UESTIONS
1. A peripheral blood film that shows increased neutrophils, c. Transformation to acute leukemia
basophils, eosinophils, and platelets is highly suggestive d. Temporary remission
of:
a. AML 5. The most common mutation found in patients with
b. CML primary PV is:
c. MDS a. BCR/ABL
d. Multiple myeloma b. Philadelphia chromosome
c. JAK2 V617F
2. Which of the following chromosome abnormalities is d. t(15;17)
associated with CML?
a. t(15;17) 6. The peripheral blood in PV typically manifests:
b. t(8;14) a. Erythrocytosis only
c. t(9;22) b. Erythrocytosis and thrombocytosis
d. Monosomy 7 c. Erythrocytosis, thrombocytosis, and granulocytosis
d. Anemia and thrombocytopenia
3. A patient has a WBC count of 30 109/L and the following
WBC differential: 7. A patient has a platelet count of 700 109/L with abnor-
Segmented neutrophils38% malities in the size, shape, and granularity of platelets; a
Bands17% WBC count of 12 109/L; hemoglobin of 11g/dL. The
Metamyelocytes7% Philadelphia chromosome is not present. The most likely
Myelocytes20% diagnosis is:
Promyelocytes10% a. PV
Eosinophils3% b. ET
Basophils5% c. CML
Which of the following test results would be helpful in d. Leukemoid reaction
determining whether the patient has CML?
a. Nitroblue tetrazolium reduction product increased 8. Complications of ET include all of the following except:
b. Myeloperoxidase increased a. Thrombosis
c. Periodic acidSchiff staining decreased b. Hemorrhage
d. LAP decreased c. Seizures
d. Infections
4. A patient in whom CML has previously been diagnosed
has circulating blasts and promyelocytes that total 30% of
leukocytes. The disease is considered to be in what phase?
a. Chronic stable phase
b. Accelerated phase
530 PART V Leukocyte Disorders

9. Which of the following patterns is characteristic of the 10. The myelofibrosis associated with PMF is a result of:
peripheral blood in patients with PMF? a. Apoptosis resistance in the fibroblasts of the bone
a. Teardrop-shaped erythrocytes, nucleated RBCs, marrow
immature granulocytes b. Impaired production of normal collagenase by the
b. Abnormal platelets only mutated cells
c. Hypochromic erythrocytes, immature granulocytes, c. Enhanced activity of fibroblasts owing to increased
and normal platelets stimulatory cytokines
d. Spherocytes, immature granulocytes, and increased d. Increased numbers of fibroblasts owing to cytokine
numbers of platelets stimulation of the pluripotential stem cells

REFERENCES
1. Fialkow PJ, Jacobson RJ, Papayannopoulou T: Chronic 18. Stam K, Heisterkamp N, Grosveld G, et al: Evidence of a new
myelocytic leukemia: clonal origin in a stem cell common chimeric bcr/c-abl mRNA in patients with chronic myelo-
to the granulocytic, erythrocytic, platelet and monocyte/ cytic leukemia and the Philadelphia chromosome. N Engl J
macrophage. Am J Med 63:125-130, 1977. Med 313:1429-1433, 1985.
2. Adamson JW, Fialkow PJ, Murphy S, et al: Polycythemia 19. Groffen J, Stephenson JR, Heisterkamp N, et al: Philadelphia
vera: stem-cell and probable clonal origin of the disease. chromosomal breakpoints are clustered within a limited
N Engl J Med 295:913-916, 1976. region, bcr, on chromosome 22. Cell 36(1):93-99, 1984.
3. Fialkow PJ, Faguet GB, Jacobson RJ, et al: Evidence that 20. Randolph TR: Chronic myelocytic leukemia part I: history,
essential thrombocythemia is a clonal disorder with origin clinical presentation, and molecular biology. Clin Lab Sci
in a multipotent stem cell. Blood 58:916-919, 1981. 18(1):38-48, 2005.
4. Jacobson RJ, Salo A, Fialkow PJ: Agnogenic myeloid meta- 21. Faderl S, Talpaz M, Estrov Z, et al: Mechanisms of disease:
plasia: a clonal proliferation of hematopoietic stem cells the biology of chronic myeloid leukemia. N Engl J Med
with secondary myelofibrosis. Blood 51:189-194, 1978. 341(3):164-172, 1999.
5. Dameshek W: Some speculations on the myeloproliferative 22. Epner DE, Koeffler HP: Molecular genetic advances in
syndromes. Blood 6:372 (editorial), 1951. chronic myelogenous leukemia. Ann Intern Med 113:3-6,
6. Vardiman JW, Thiele J, Arber DA, et al: The 2008 revision of 1990.
the World Health Organization (WHO) classification of 23. Douer D, Levine AM, Sparkes RS, et al: Chronic myelocytic
myeloid neoplasms and acute leukemia: rationale and leukemia: a pluripotent haemopoietic cell is involved in the
important changes. Blood 114: 937-951, 2009. malignant clone. Br J Haematol 49:615-619, 1981.
7. Whang J, Frei E III, Tijo JH, et al: The distribution of the 24. Bhatia R, Verfaillie CM: The effects of interferon-alpha on
Philadelphia chromosome in patients with chronic myelog- beta-1 integrin mediated adhesion and growth regulation in
enous leukemia. Blood 22:664-673, 1963. chronic myelogenous leukemia. Leuk Lymphoma 28:241-
8. Spiers AS, Bain BJ, Turner JE: The peripheral blood 254, 1998.
in chronic granulocytic leukemia: study of 50 untreated 25. Georgii A, Buesche G, Krept A: The histopathology of
Philadelphia-positive cases. Scand J Hematol 18:25-28, chronic myeloproliferative disorders: review. Ballieres Clin
1977. Haematol 11:721-749, 1998.
9. Ward HP, Block MH: The natural history of agnogenic 26. Klineberg JR, Bluestone R, Schlosstein L, et al: Urate deposi-
myeloid metaplasia (AMM) and a critical evaluation of its tion disease: how it is regulated and how can it be modified?
relationship with the myeloproliferative syndrome. Medicine Ann Intern Med 78:99-111, 1973.
(Baltimore) 150:357-420, 1971. 27. Lichtman MA, Rowe JM: Hyperleukocytic leukemias:
10. Mitus AJ, Schafer AI: Thrombocytosis and thrombocythe- rheological, clinical and therapeutic considerations. Blood
mia. Hematol Oncol Clin North Am 4:157-178, 1990. 60:279-283, 1982.
11. Gilbert HS: Familial myeloproliferative disease. Ballieres 28. Lichtman MA, Heal J, Rowe JM: Hyperleukocytic leukemia.
Clin Haematol 11:849-858, 1998. Ballieres Clin Haematol 1:725-746, 1987.
12. Barr RD, Fialkow PJ: Clonal origin of chronic myelocytic 29. Muehleck SD, McKenna RD, Arthur DC, et al: Transforma-
leukemia. N Engl J Med 289:307-309, 1973. tion of chronic myelogenous leukemia: clinical, morpho-
13. Bizzozzero OJ, Johnson KG, Ciocco A: Radiation-related logic and cytogenetic features. Am J Clin Pathol 82:1-14, 1984.
leukemia in Hiroshima and Nagasaki, 1946-1964: I. Distri- 30. Vardiman JW, Melo JV, Baccarani M, et al: Chronic myelog-
bution, incidence, and appearance time. N Engl J Med enous leukemia., BCR-ABL1 positive. In Swerdlow SH,
274:1095-1101, 1966. Campo E, Harris NL, et al, editors: WHO classification of
14. Brown WM, Doll R: Mortality from cancer and other causes tumours of haematopoietic and lymphoid tissues, ed 4, Lyon,
after radiotherapy for ankylosing spondylitis. BMJ France, 2008, IARC Press, pp 32-37.
5474:1327-1332, 1965. 31. Bernstein R: Cytogenetics of chronic myelogenous leuke-
15. Marmont A, Frassoni F, Bacigalupo A, et al: Recurrence of mia. Semin Hematol 25:20-34, 1988.
Ph1-positive leukemia in donor cells after marrow trans- 32. Sandberg AA: Chromosomes in the chronic phase of CML.
plantation for chronic granulocytic leukemia. N Engl J Med Virchows Arch B Cell Pathol 29:51-55, 1978.
310:903-906, 1984. 33. Bareford D, Jacobs P: Chronic neutrophilic leukemia. Am J
16. Nowell P, Hungerford DA: A minute chromosome in human Clin Pathol 73:837, 1980.
granulocytic leukemia. Science 132:1497-1499, 1960. 34. Bearman RM, Kjeldsburg CR, Pangalis GA, et al: Chronic
17. Rowley JD: A new consistent chromosome abnormality in monocytic leukemia in adults. Cancer 48:2239-2255, 1981.
chronic myelogenous leukemia identified by quinacrine 35. Thomas WJ, North RB, Poplack DG, et al: Chronic myelo-
fluorescence and Giemsa banding. Nature 243:290-293, monocytic leukemia in childhood. Am J Hematol 10:181-
1973. 194, 1981.
CHAPTER 34 Myeloproliferative Neoplasms 531
36. Mijovic A, Mufti GJ: The myelodysplastic syndromes: 58. Berlin N: Diagnosis and classification of the polycythemias.
towards a functional classification. Blood Rev 12:73-83, Semin Hematol 12:339-351, 1975.
1998. 59. Berk PD, Goldberg JN, Donovan PB, et al: Therapeutic rec-
37. Kurzrock R, Gutterman JU, Talpaz M: The molecular genet- ommendations in polycythemia vera based on Polycythe-
ics of Philadelphia chromosome positive leukemias. N Engl mia Vera Study Group protocols. Semin Hematol 23:132-143,
J Med 319:990-998, 1988. 1986.
38. Specchia G, Mininni D, Guerrasio A, et al: Ph positive acute 60. Wolf BC, Bank PM, Mann RB, et al: Splenic hematopoiesis
lymphoblastic leukemia in adults: molecular and clinical in polycythemia vera: a morphologic and immunohisto-
studies. Leuk Lymphoma 18(suppl):37-42, 1995. logic study. Am J Clin Pathol 89:69-75, 1988.
39. Faderl S, Talpaz M, Estrov A, et al: Chronic myelogenous 61. Ellis JT, Peterson P, Geller SA, et al: Studies of the bone
leukemia: biology and therapy. Ann Intern Med 131:207- marrow in polycythemia vera and the evolution of myelo-
219, 1999. fibrosis and second hematologic malignancies. Semin
40. Catovsky D: Ph1-positive acute leukemia and chronic granu- Hematol 23:144-155, 1986.
locytic leukemia: one or two diseases? Br J Haematol 42:493- 62. Mitus AJ, Schafer A: Thrombocytosis and thrombocythemia.
498, 1979. Hematol Oncol Clin North Am 4:157-178, 1990.
41. Wintrobe MM, Huguley CM, McLennan MT, et al: Nitrogen 63. Thiele J, Kvasnicka HM, Orazi A, et al: Essential thrombocy-
mustard as a therapeutic agent for Hodgkins disease, lym- themia. In Swerdlow SH, Campo E, Harris NL, et al, editors:
phosarcoma and leukemia. Ann Intern Med 27:529-539, WHO classification of tumours of haematopoietic and lymphoid
1947. tissues, ed 4, Lyon, France, 2008, IARC Press, pp 48-53.
42. Galton DA: The use of myleran in chronic myeloid leuke- 64. Murphy S, Iland H, Rosenthal D, et al: Essential thrombo-
mia: results of treatment. Lancet 264:208-213, 1953. cythemia: an interim report from the Polycythemia Vera
43. Lion T, Gaiger A, Henn T, et al: Use of quantitative poly- Study Group. Semin Hematol 23:177-182, 1986.
merase chain reaction to monitor residual disease in chronic 65. Campbell PJ, Scott LM, Buck G, et al: Definitions of sub-
myelogenous leukemia during treatment with interferon. types of essential thrombocythemia and relation to polycy-
Leukemia 9:1353-1360, 1995. themia vera based on JAK2 V617F mutation status: a
44. Sawyers CL: Chronic myeloid leukemia. N Engl J Med prospective study. Lancet 366:1945-1953, 2005.
340:1330-1340, 1999. 66. Vannucchi AM, Antonioli E, Guglielmelli P, et al: Charac-
45. Guilhot F, Chastang C, Michallet M, et al: Interferon alpha teristics and clinical correlates of MPL 515W>L/K mutation
2b combined with cytarabine versus interferon alone in in essential thrombocythemia. Blood 112:844-847, 2008.
chronic myelogenous leukemia. N Engl J Med 337:223-229, 67. Tefferi A, Vardiman JW: Classification and diagnosis of
1997. myeloproliferative neoplasms: the 2008 World Health
46. OBrien SG: Autografting for chronic myeloid leukemia. Organization criteria and point-of-care diagnostic algo-
Baillieres Clin Haematol 10:369-388, 1997. rithms. Leukemia 22:14-22, 2008.
47. Atwell S, Adams JM, Badger J, et al: A novel mode of Gleevec 68. Kuecht H, Streuli RA: Megakaryopoiesis in different forms
binding is revealed by the structure of spleen tyrosine of thrombocytosis and thrombocytopenia: identification of
kinase. J Biol Chem 279(53):55827-55832, 2004. megakaryocyte precursors by immunostaining of intracyto-
48. Druker BJ, Talpaz M, Resta DJ, et al: Efficacy and safety of a plasmic factor VIII-related antigen. Acta Haematol 74:208-
specific inhibitor of the BCR-ABL tyrosine kinase in chronic 212, 1985.
myeloid leukemia. N Engl J Med 344:1031-1037, 2001. 69. McCarthy DM: Fibrosis of the bone marrow, content and
49. Lin F, Drummond M, OBrien S, et al: Molecular monitoring causes. Br J Haematol 59:1-7 (annotation), 1985.
in chronic myeloid leukemia patients who achieve complete 70. Reilly IF: Pathogenesis of idiopathic myelofibrosis: present
remission on imatinib. Blood 102:1143 (letter), 2003. status and future directions. Br J Haematol 88:1-8, 1994.
50. Nardi A, Azam M, Daley GO: Mechanisms and implications 71. Wang JC, Cheung CP, Fakhiuddin A, et al: Circulating granu-
of imatinib resistance mutations in BCR-ABL. Curr Opin locyte and macrophage progenitor cells in primary and sec-
Hematol 11:35-43, 2004. ondary myelofibrosis. Br J Haematol 54:301-307, 1983.
51. Kantarjian HM, Talpaz M, Giles F, et al: New insights into 72. Jacobson RJ, Salo A, Fialkow PJ: Agnogenic myeloid meta-
the pathophysiology of chronic myeloid leukemia and ima- plasia: a clonal proliferation of hematopoietic stem cells
tinib resistance. Ann Intern Med 145:913-923, 2006. with secondary myelofibrosis. Blood 51:189-194, 1978.
52. Gilliland DG, Blanchard KL, Levy J, et al: Clonality in 73. Buschle M, Janssen JY, Drexler H, et al: Evidence for pluripo-
myeloproliferative disorders: analysis by means of the poly- tent stem cell origin of idiopathic myelofibrosis: clonal
merase chain reaction. Proc Natl Acad Sci U S A 88:6848- analysis of a case characterized by a N-ras gene mutation.
6852, 1991. Leukemia 2:658-660, 1988.
53. Prchal JF, Adamson JW, Murphy S, et al: Polycythemia vera: 74. Juvonen E: Megakaryocyte colony formation in chronic
the in vitro response of normal and abnormal stem cell lines myeloid leukemia and myelofibrosis. Leuk Res 12:751-756,
to erythropoietin. J Clin Invest 61:1044-1047, 1978. 1988.
54. Adamson JW, Singer JW, Catalano P, et al: Polycythemia 75. Barosi G, Viarengo G, Pecci A, et al: Diagnostic and clinical
vera: further in vitro studies of hematopoietic regulation. relevance of the number of circulating CD34(+) cells in
J Clin Invest 66:1363-1368, 1980. myelofibrosis with myeloid metaplasia. Blood 98:3249-
55. James C, Ugo V, Le Couedic JP, et al: A unique clonal JAK2 3255, 2001.
mutation leading to constitutive signalling causes polycy- 76. Jacobs P, Maze S, Tayob F, et al: Myelofibrosis, splenomegaly,
themia vera. Nature 434:1144-1148, 2005. and portal hypertension. Acta Haematol 74:45-48, 1985.
56. Baxter EJ, Scott LM, Campbell PJ, et al: Acquired mutation 77. Vellenga E, Mulder N, The T, et al: A study of the cellular
of the tyrosine kinase JAK2 in human myeloproliferative and humoral immune response in patients with myelofibro-
disorders. Lancet 365:1054-1061, 2005. sis. Clin Lab Haematol 4:239-246, 1982.
57. Levine RL, Wadleigh M, Cools J, et al: Activating mutation 78. Pegrum GD, Foadi M, Boots M, et al: How should we
in the tyrosine kinase JAK2 in polycythemia vera, essential manage myelofibrosis? J R Coll Physicians Lond 15:17-18,
thrombocythemia, and myeloid metaplasia with myelofi- 1981.
brosis. Cancer Cell 7:387-397, 2005. 79. Tefferi A, Silverstein MN: Current perspective in agnogenic
532 PART V Leukocyte Disorders

myeloid metaplasia. Leuk Lymphoma 22(suppl 1):169-171, 1380, 2006.


1996. 92. Hussein K, Bock O, Seegers A, et al: Myelofibrosis
80. Guardiola P, Anderson JE, Bandini G, et al: Allogeneic stem evolving during imatinib treatment of chronic myeloprolif-
cell transplantation for agnogenic myeloid metaplasia: a erative disease with coexisting BCR/ABL translocation and
European Group for Blood and Marrow Transplantation, JAK2V617F mutation. Blood 109:4106-4107, 2007.
Socit Franaise de Greffe de Moelle, Gruppo Italiano per 93. Kramer A, Reiter A, Kruth J, et al: JAK2-V617F mutation in
il Trapianto del Midollo Osseo, and Fred Hutchinson a patient with Philadelphia-chromosome positive chronic
Cancer Research Center Collaborative Study. Blood 93:2831- myeloid leukemia. Lancet Oncol 8:658-660, 2007.
2838, 1999. 94. Pardanani AD, Levine RL, Lasho T, et al: MPL515 mutations
81. Xu M, Bruno E, Chao J, et al: The constitutive mobilization in myeloproliferative and other myeloid disorders: a study
of bone marrowrepopulating cells into the peripheral of 1182 patients. Blood 108:3472-3476, 2006.
blood in idiopathic myelofibrosis. Blood 105:1699-1705, 95. Li S, Kralovics R, De Libero G, et al: Clonal heterogeneity in
2005. polycythemia vera patients with JAK2 exon 21 and JAK2-
82. Ishii T, Zhao Y, Sozer S, et al: Behavior of CD34+ cells iso- V617F mutations. Blood 111:3863-3866, 2008.
lated from patients with polycythemia vera in NOD/SCID 96. Petra D, Li S, Brisci A, et al: Somatic mutations of JAK2 exon
mice. Exp Hematol 35:1633-1640, 2007. 12 in patients with JAK2 (V617F)negative myeloprolifera-
83. James C, Mazurier F, Dupont S, et al: The hematopoietic tive disorders. Blood 111:1686-1689, 2008.
stem cell compartment of JAK2V617F-positive myeloprolif- 97. Kralovics R, Stockton D, Prchal J, et al: Clonal hematopiesis
erative disorders is a reflection of disease heterogeneity. in familial polycythemia vera suggests the involvement of
Blood 112:2429-2438, 2008. multiple mutational events in the early pathogenesis of the
84. Pardanani A, Fridley BL, Lash TL, et al: Host genetic varia- disease. Blood 102:3793-3796, 2003.
tion contributes to phenotypic diversity in myeloprolifera- 98. Bellanne-Chantelot C, Chaumarel I, Labopin M, et al:
tive disorders. Blood 111:2785-2789, 2008. Genetic and clinical implications of the Val617Phe JAK2
85. Dupont S, Masse A, James C, et al: The JAK2 617 V>F muta- mutation in 72 families with myeloproliferative disorders.
tion triggers erythrocyte hypersensitivity and terminal ery- Blood 108:346-352, 2006.
throid amplification in primary erythroid cells from patients 99. Villeval JL, Cohen-Solal K, Tulliez M, et al: High thrombo-
with polycythemia vera. Blood 110:1013-1021, 2007. poietin production by hematopoietic cells induces a fatal
86. Scott LM, Scott MA, Campbell PJ, et al: Progenitors homo- myeloproliferative syndrome in mice. Blood 90:4369-4383,
zygous for the V617F mutation occur in most patients with 1997.
polycythemia vera but not essential thrombocythemia. 100. Bain BJ, Brunning RD, Vardiman JW, et al: Chronic neutro-
Blood 108:2435-2437, 2006. philic leukemia. In Swerdlow SH, Campo E, Harris NL,
87. Xing S, Wanting TH, Zhao W, et al: Transgenic expression of et al, editors: WHO classification of tumours of haematopoietic
JAK2V617F causes myeloproliferative disorders in mice. and lymphoid tissues, ed 4, Lyon, France, 2008, IARC Press,
Blood 111:5109-5117, 2008. pp 38-39.
88. Tiedt R, Hao-Shin S, Sobas MA, et al: Ratio of mutant JAK2- 101. Bain BJ, Gilliland DG, Vardiman JW, et al: Chronic eosino-
V617F to wild-type JAK2 determines the MPD phenotype in philic leukaemia, not otherwise specified. In Swerdlow SH,
transgenic mice. Blood 111:3931-3940, 2008. Campo E, Harris NL, et al, editors: WHO classification of
89. Kiladjian, JJ, Elkassar N, Cassinat B, et al: Essential thrombo tumours of haematopoietic and lymphoid tissues, ed 4, Lyon,
cythemias without V617F JAK2 mutation are clonal hema- France, 2008, IARC Press, pp 51-53.
topoietic stem cell disorders. Leukemia 20:1181-1183, 2006. 102. Horny HP, Metcalfe DD, Bennett JM, et al: Mastocytosis. In
90. Levine RL, Belisle C, Wadleigh M, et al: X-inactivation-based Swerdlow SH, Campo E, Harris NL, et al, editors: WHO
clonality analysis and quantitative JAK2V617F assessment classification of tumours of haematopoietic and lymphoid tissues,
reveal a strong association between clonality and JAK2V617F ed 4, Lyon, France, 2008, IARC Press, pp 54-63.
in PV but not ET/MMM, and identifies a subset of 103. Kvasnicka HM, Bain BJ, Thiele J, et al: Myeloproliferative
JAK2V617F-negative ET and MMM patients with clonal neoplasm, unclassifiable. In Swerdlow SH, Campo E, Harris
hematopoiesis. Blood 107:4139-4141, 2006. NL, et al, editors: WHO classification of tumours of haemato-
91. Kralovics R, Teo SS, Li SS, et al: Acquisition of the V617F poietic and lymphoid tissues, ed 4, Lyon, France, 2008, IARC
mutation of JAK2 is a late genetic event in a subset of Press, pp 64-65.
patients with myeloproliferative disorders. Blood 108:1377-
Myelodysplastic Syndromes
Bernadette F. Rodak
35
OUTLINE OBJECTIVES
Etiology After completion of this chapter, the reader will be able to:
Morphologic Abnormalities
in Peripheral Blood and 1. Define myelodysplastic syndromes (MDSs). 7. Discuss prognostic indicators in MDS.
Bone Marrow 2. Explain the etiology of MDS. 8. Discuss modes of management for MDS.
Dyserythropoiesis 3. Recognize morphologic features of dyspoiesis in 9. Discuss the epidemiology of MDS and apply it as a
Dysmyelopoiesis bone marrow and peripheral blood. contributor in differential diagnosis.
Dysmegakaryopoiesis 4. Discuss abnormal functions of granulocytes, eryth- 10. Suggest laboratory tests and their results that would
Differential Diagnosis rocytes, and thrombocytes in MDS. rule out MDS in the differential diagnosis.
Abnormal Cellular 5. Correlate peripheral blood, bone marrow, and 11. Explain the rationale for the category
Function cytogenetic findings in MDS with classification of myelodysplastic/myeloproliferative neoplasms
Classification of systems. (MDS/MPN).
Myelodysplastic
6. Compare and contrast the French-American-British 12. Correlate peripheral blood, bone marrow, and
Syndromes
and the 2008 World Health Organization classifica- cytogenetic findings in MDS/MPN with disease
French-American-British
Classification tions of MDS. classification.
World Health Organization
Classification CASE STUDY
Myelodysplastic/
Myeloproliferative After studying the material in this chapter, the reader should be able to respond to the following
Neoplasms case study:
Chronic Myelomonocytic
A 43-year-old man experienced fatigue and malaise. He had pancytopenia (WBC count of 2.2
Leukemia
Atypical Chronic Myeloid 109/L, hemoglobin of 6.1g/dL, platelet count of 51 109/L). The WBC differential was essentially
Leukemia, BCR/ABL1 normal. Mean cell volume was 132fL (reference range, 80 to 100fL), and vitamin B12 and folate
Negative levels were normal. The bone marrow was normocellular with a myeloid-to-erythroid ratio of 1:1
Juvenile Myelomonocytic and adequate megakaryocytes. The erythroid component was dysplastic with megaloblastic features.
Leukemia No abnormal localization of immature precursors was noted. Chromosome analysis indicated
Myelodysplastic/ direct duplication of chromosome 1q. The patient was maintained with transfusions over the next
Myeloproliferative 6 years. At that time, his bone marrow revealed increased erythropoiesis, decreased granulopoiesis,
Neoplasm, and megakaryopoiesis, all with dysplastic changes. There were 50% to 60% ringed sideroblasts.
Unclassifiable 1. What should be included in the differential diagnosis of patients with pancytopenia and elevated
Cytogenetics and
mean cell volume?
Molecular Genetics
2. Given the normal vitamin B12 and folate levels, what is the patients probable diagnosis?
Cytogenetics
Molecular Alterations 3. In which WHO classification does this disorder belong?
Prognosis
Treatment
Future Directions

F
or decades, laboratory professionals have observed a Historically, this pattern of abnormalities was referred to
group of morphologic abnormalities in peripheral blood as refractory anemia, smoldering leukemia, oligoblastic leukemia,
films and bone marrow smears of elderly patients. The or preleukemia.1-3 In 1982, the French-American-British (FAB)
findings were heterogeneous and affected all cell lines, and the Cooperative Leukemia Study Group proposed terminology and
condition either remained stable for years or progressed rapidly a specific set of morphologic criteria to describe what are now
to death. known as myelodysplastic syndromes (MDSs).4 In 1997, a group

533
534 PART V Leukocyte Disorders

from the World Health Organization (WHO) proposed a patients with MDS have increased levels of angiogenic growth
new classification that included molecular, cytogenetic, and factors, including vascular endothelial growth factor.31,32
immunologic criteria in addition to morphologic features.5,6 Therapy-related MDS occurs in patients who have been
The WHO classification was revised in 2008. Both the FAB treated previously with chemotherapy or radiotherapy or both.
and WHO classifications are discussed in this chapter. Median onset of therapy-related MDS varies with the agents
MDSs are a group of acquired clonal hematologic disorders used and is usually 2.5 to 5.0 years after therapy was initi-
characterized by progressive cytopenias in the peripheral ated,33,34 although cases occurring 7 years after initial exposure
blood, reflecting defects in erythroid, myeloid, and/or mega- to chemotherapy or radiation have been reported.35 Therapy-
karyocytic maturation.7,8 The median age at diagnosis is 70. related MDS often is more aggressive and may evolve quickly
MDS rarely affect individuals younger than age 50 unless pre- into acute myeloblastic leukemia (AML).20,34,35 The 2008 WHO
ceded by chemotherapy or radiation for another malignancy.1,9 classification places therapy-related MDSs into the AML cate-
Cases in young adults and children have been reported, gory of therapy-related myeloid neoplasms (see Chapter 36).
however.10,11 The incidence of these disorders seems to be
increasing, but this apparent increase may be attributable in
MORPHOLOGIC ABNORMALITIES IN
part to improved techniques for identifying these diseases and
PERIPHERAL BLOOD AND BONE MARROW
to improved classification.12,13 At this time, the fastest-growing
segment of the population is the group older than 60 years of In MDS each of the three major myeloid cell lines has dyspoi-
age. MDSs are becoming a more common finding in the hema- etic morphologic features. The following sections provide
tology laboratory, and familiarity with these disorders is an descriptions of common abnormal morphologic findings.4,9,19
essential part of the body of knowledge of all medical labora- These descriptions are not all inclusive because of the large
tory professionals. number of possible cellular mutations and combinations of
mutations.

ETIOLOGY
Dyserythropoiesis
MDS may arise de novo (primary MDS) or as a result of therapy In the peripheral blood, the most common morphologic
(therapy-related MDS). Although MDSs are a group of hetero- finding in dyserythropoiesis is the presence of oval macrocytes
geneous diseases, all are the result of proliferation of abnormal (Figure 35-1). When these cells are seen in the presence of
stem cells.7,8,14,15 The initiating defect in most cases is at the normal vitamin B12 and folate values, MDS should be included
level of the myeloid stem cell, because primarily the erythroid, in the differential diagnosis. Hypochromic microcytes in the
myeloid, and megakaryocytic cells are affected. It may be that presence of adequate iron stores also are seen in MDS. A
the affected hematopoietic stem cell has lost its lymphopoietic dimorphic red blood cell (RBC) population (Figure 35-2) is
potential, because only rarely does MDS transform to acute another indication of the clonality of this disease. Poikilocyto-
lymphoblastic leukemia.16,17 The abnormal stem cell may be sis, basophilic stippling, Howell-Jolly bodies, and siderocytes
the result of the cumulative effects of environmental exposure also are indications that the erythrocyte has undergone abnor-
in susceptible individuals. Mutations may be caused by chemi- mal development.36
cal insult, radiation, or viral infection. There also may be an Dyserythropoiesis in the bone marrow is evidenced by
association with smoking.18 An association with inherited RBC precursors with more than one nucleus or abnormal
hematologic disorders has also be found.19 The mutated stem nuclear shapes. The normally round nucleus may have lobes
cell produces a pathologic clone of cells that expands in size or buds. Nuclear fragments may be present in the cytoplasm
at the expense of normal cell production.18,20 Because each (Figure 35-3). Internuclear bridging is occasionally present
mutation produces a unique clone with a specific cellular
defect, MDSs have a multitude of expressions. Two morpho-
logic findings are common to all types of MDS, however: the
presence of progressive cytopenias despite cellular bone
marrows and dyspoiesis in one or more cell lines.
Disruption of apoptosis may be responsible for the ineffec-
tive hematopoiesis in MDS.21-26 Apoptosis (programmed cell
death) regulates cell population by decreasing cell survival. In
MDS, apoptosis is increased in early disease, when peripheral
blood cytopenias are evident. Later in MDS, when progression
toward leukemia is apparent, apoptosis has been shown to be
decreased, which allows increased neoplastic cell survival and
expansion of the abnormal clone.27-30 Other important factors
include the levels of antiangiogenic cytokines, tumor necrosis
factor, and cellular components of the immune system, as well
as the interaction between MDS clonal cells and the hemato-
poietic inductive microenvironment. Evidence has shown that Figure 35-1 Oval macrocytes in peripheral blood (1000).
CHAPTER 35 Myelodysplastic Syndromes 535

Figure 35-2 Dimorphic erythrocyte population, including macrocytic and Figure 35-5 Bone marrow specimen showing heterogeneous staining in
microcytic cells (in peripheral blood, 500). a bilobed erythroid precursor (1000).

Figure 35-3 Bone marrow specimen showing erythroid hyperplasia and Figure 35-6 This myelocyte (right) in peripheral blood has a nucleus with
nuclear budding in erythroid precursors (1000). clumped chromatin and a basophilic immature cytoplasm showing asyn-
chrony. Note also the agranular myeloid cell (left) (1000).

BOX 35-1 Morphologic Evidence of Dyserythropoiesis


Oval macrocytes
Hypochromic microcytes
Dimorphic red blood cell (RBC) population
RBC precursors with more than one nucleus
RBC precursors with abnormal nuclear shapes
RBC precursors with uneven cytoplasmic staining
Ringed sideroblasts

in these cases may have erythrocytic hyperplasia or hypoplasia


(Box 35-1).
Figure 35-4 Erythroid precursors showing nuclear bridging (arrow) (bone
marrow, 1000). Dysmyelopoiesis
Dysmyelopoiesis in the peripheral blood is suspected when
(Figure 35-4).37 Abnormal cytoplasmic features may include there is a persistence of basophilia in the cytoplasm of other-
basophilic stippling or heterogeneous staining (Figure 35-5). wise mature white blood cells (WBCs), indicating nuclear-
Ringed sideroblasts are a common finding. Megaloblastoid cel- cytoplasmic asynchrony (Figure 35-6). Abnormal granulation
lular development in the presence of normal vitamin B12 and of the cytoplasm of neutrophils, in the form of larger than
folate values is another indication of MDS. The bone marrows normal granules, hypogranulation, or the absence of granules,
536 PART V Leukocyte Disorders

Figure 35-7 Agranular myeloid cells (peripheral blood, 1000). Figure 35-9 Uneven staining of white blood cell cytoplasm (bone marrow,
1000).

Figure 35-8 Nuclear ring in myeloid cell (peripheral blood, 1000). Figure 35-10 Promyelocyte or myelocyte devoid of granules and an
agranular neutrophil (bone marrow, 1000).

is a common finding. Agranular bands can be easily misclassi-


fied as monocytes (Figure 35-7). Abnormal nuclear features BOX 35-2 Morphologic Evidence of Dysmyelopoiesis
may include hyposegmentation, hypersegmentation, or nuclear Persistent basophilic cytoplasm
rings (Figure 35-8).38 Abnormal granulation
In the bone marrow, dysmyelopoiesis may be represented Abnormal nuclear shapes
by nuclear-cytoplasmic asynchrony. Cytoplasmic changes Uneven cytoplasmic staining
include uneven staining, such as a dense ring of basophilia
around the periphery with a clear unstained area around the
nucleus or whole sections of cytoplasm unstained with the Abnormal localization of immature precursors is a charac-
remainder of the cytoplasm stained normally (Figure 35-9). teristic finding in bone marrow biopsy specimens from patients
There may be abnormal granulation of the cytoplasm in which with MDS.41 Normally, myeloblasts and promyelocytes reside
promyelocytes or myelocytes or both are devoid of primary along the endosteal surface of the bone marrow. In some
granules (Figure 35-10), primary granules may be larger than cases of MDS, these cells tend to cluster centrally in marrow
normal, or secondary granules may be reduced in number or sections.
absent, and there may be an occasional Auer rod.39,40 Agranular
promyelocytes may be mistaken for blasts; this could lead to Dysmegakaryopoiesis
misclassification of the disease in the AML scheme. Abnormal Platelets also exhibit dyspoietic morphology in the peripheral
nuclear findings may include hypersegmentation or hyposeg- blood. Common changes include giant platelets and abnormal
mentation and possibly ring-shaped nuclei (Box 35-2). platelet granulation, either hypogranulation or agranulation
The bone marrow may exhibit granulocytic hypoplasia or (Figure 35-11). Some platelets may possess large fused
hyperplasia. Monocytic hyperplasia is a common finding in granules. Circulating micromegakaryocytes may be present in
dysplastic marrows. peripheral blood from patients with MDS (Figure 35-12).9
CHAPTER 35 Myelodysplastic Syndromes 537

BOX 35-3 Morphologic Evidence of


Dysmegakaryopoiesis
Giant platelets
Platelets with abnormal granulation
Circulating micromegakaryocytes
Large mononuclear megakaryocytes
Micromegakaryocytes or micromegakaryoblasts or both
Abnormal nuclear shapes in the megakaryocytes/blasts

DIFFERENTIAL DIAGNOSIS
Dysplasia by itself is not sufficient evidence for MDS, because
Figure 35-11 Abnormal platelet granulation (arrow) (peripheral blood, several other conditions can cause similar morphologic fea-
1000). tures. Some examples are vitamin B12 or folate deficiency,
which can cause pancytopenia and dysplasia, and exposure to
heavy metals. Copper deficiency may cause reversible myelo-
dysplasia.42 Some congenital hematologic disorders, such as
Fanconi anemia and congenital dyserythropoietic anemia, may
also present with dysplasia. Parvovirus B19 and some chemo-
therapeutic agents may give rise to dysplasia similar to that in
MDS. Paroxysmal nocturnal hemoglobinuria has similar fea-
tures. Therefore a thorough history and physical examination,
including questioning about exposure to drugs and chemicals,
is essential.9

ABNORMAL CELLULAR FUNCTION


The cells produced by abnormal maturation not only have an
abnormal appearance but also have abnormal function.9,43 The
granulocytes may have decreased adhesion,44,45 deficient phago-
Figure 35-12 Micromegakaryocyte (peripheral blood, 1000).
cytosis,45 decreased chemotaxis,44,45 or impaired microbicidal
capacity.46 Decreased levels of myeloperoxidase and alkaline
phosphatase may be found.47 The RBCs may exhibit shortened
survival,48 and erythroid precursors may have a decreased
response to erythropoietin that may contribute to anemia.49
Patients may experience increased bleeding despite adequate
platelet numbers.9,50,51 The type and degree of dysfunction
depend on the mutation present in the hematopoietic stem cell.

CLASSIFICATION OF
MYELODYSPLASTIC SYNDROMES
French-American-British Classification
In an effort to standardize the diagnosis of MDSs, the FAB
created five classes of MDS, each with a specific set of morpho-
logic criteria. The categories were defined by the amount of
dysplasia and the number of blasts in the bone marrow. The
Figure 35-13 Megakaryocyte with small separated nuclei (bone marrow,
1000). diagnosis of acute leukemia required at least 30% blasts in the
bone marrow.4 The FAB classification included the following:
1. Refractory anemia
The megakaryocytic component of the bone marrow may 2. Refractory anemia with ringed sideroblasts (RARS)
exhibit abnormal morphology: large mononuclear megakaryo- 3. Refractory anemia with excess blasts (RAEB)
cytes, micromegakaryocytes, or micromegakaryoblasts. The 4. Chronic myelomonocytic leukemia (CMML)
nuclei in these cells may be bilobed or have multiple small, 5. Refractory anemia with excess blasts in transformation
separated nuclei (Figure 35-13; Box 35-3).9 (RAEB-t)
538 PART V Leukocyte Disorders

TABLE 35-1 Peripheral Blood and Bone Marrow Findings in Myelodysplastic Syndromes (MDSs)
Disease Blood Findings Bone Marrow Findings
Refractory cytopenia with unilineage dysplasia (RCUD); Unicytopenia* Unilineage dysplasia: 10% of cells in one myeloid lineage
refractory anemia (RA); refractory neutropenia (RN); No or rare blasts (<1%) <5% blasts
refractory thrombocytopenia (RT) <15% of erythroid precursors are ringed sideroblasts
Refractory anemia with ringed sideroblasts (RARS) Anemia 15% of erythroid precursors are ringed sideroblasts
No blasts Erythroid dysplasia only
<5% blasts
Refractory cytopenia with multilineage dysplasia Cytopenia(s) Dysplasia in 10% of cells in two or more myeloid lineages
(RCMD) No or rare blasts (<1%) (neutrophil and/or erythroid precursors and/or megakaryocytes)
No Auer rods <5% blasts in marrow
<1 109/L monocytes No Auer rods
15% ringed sideroblasts
Refractory anemia with excess blasts 1 (RAEB-1) Cytopenia(s) Unilineage or multilineage dysplasia
<5% blasts 5-9% blasts
No Auer rods No Auer rods
<1 109/L monocytes
Refractory anemia with excess blasts 2 (RAEB-2) Cytopenia(s) Unilineage or multilineage dysplasia
5-19% blasts 10-19% blasts
Auer rods Auer rods
<1 109/L monocytes
Myelodysplastic syndrome, unclassified (MDS-U) Cytopenia(s) Unequivocal dysplasia in <10% of cells in one or more myeloid
1% blasts cell lines when accompanied by a cytogenetic abnormality
considered as presumptive evidence for a diagnosis of MDS
<5% blasts
MDS associated with isolated del(5q) Anemia Normal to increased megakaryocytes with hypolobulated nuclei
Usually normal or increased <5% blasts
platelet count Isolated del(5q) cytogenetic abnormality
No or rare blasts (<1%) No Auer rods

From Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classification of tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008, IARC Press.
*Bicytopenia may occasionally be observed. Cases with pancytopenia should be classified as MDS-U.

If the marrow myeloblast percentage is <5% but there are 2-4% myeloblasts in the blood, the diagnostic classification is RAEB-1. Cases of RCUD and RCMD with 1% myeloblasts
in the blood should be classified as MDS-U.

Cases with Auer rods and <5% myeloblasts in the blood and <10% myeloblasts in the marrow should be classified as RAEB-2.

The FAB classification provided a framework for discussion


of a seemingly heterogeneous group of disorders; however, its BOX 35-4 World Health Organization Classification
reliance on morphology alone limited its usefulness as a prog- of Myelodysplastic Syndromes (2008)
nostic indicator. In addition, the FAB classification did not view Refractory cytopenia with unilineage dysplasia
MDSs in their totality, because it did not address therapy-related Refractory anemia with ringed sideroblasts
or hereditary forms, and the special case of childhood MDS Refractory cytopenia with multilineage dysplasia
was not considered. Advances in medical knowledge, including Refractory anemia with excess blasts
molecular analysis, have allowed integration of clinical, immu- Myelodysplastic syndrome with isolated del(5q)
nologic, genetic, and molecular data with morphologic features. Myelodysplastic syndrome, unclassifiable
The WHO classification retains many of the FAB features while Childhood myelodysplastic syndrome (provisional)
recognizing molecular, cytogenetic, and immunologic charac- From Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classification of
teristics of these disorders. The WHO classification also removed tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
the problematic categories of CMML and RAEB-t and placed IARC Press.
them in MDS/MPD and acute leukemia, respectively.19,52
the WHO criteria added the category of refractory cytopenia
World Health Organization Classification with unilineage dysplasia (RCUD), refined some categories,
The original modifications from the FAB classification of MDS and added the provisional category of childhood MDS, also
included a reduction in the percentage of blasts required for called refractory cytopenia of childhood. The 2008 WHO classifi-
diagnosis of AML from 30% to 20% and the recognition of two cation is outlined in Box 35-4 and detailed in Table 35-1. The
new classifications: refractory cytopenia with multilineage dys- classification is extensive, and only the highlights are presented
plasia (RCMD) and del(5q) syndrome. The 2008 revision of in this chapter.19
CHAPTER 35 Myelodysplastic Syndromes 539
Refractory Anemia with Excess Blasts
Trilineage cytopenias, as well as significant dysmyelopoiesis,
dysmegakaryopoiesis, or both, are common in RAEB. Accord-
ing to the WHO classification, the peripheral blood must
contain 2% to 19% blasts. In the bone marrow, blasts number
5% to 19%. RAEB is distinguished from RCMD by myeloblast
percentage. Because there are significant differences in survival
and evolution to AML may occur, the WHO classification
divided RAEB into two types depending on the percentage of
blasts in blood and bone marrow:
RAEB-15% to 9% blasts in the bone marrow or 2% to 4%
blasts in the peripheral blood
RAEB-210% to 19% blasts in the bone marrow and 5% to
19% blasts in the peripheral blood
Figure 35-14 Ringed sideroblast (arrows) (bone marrow, Prussian blue The presence of Auer rods, regardless of blast count, qualifies
stain, 1000). a case as RAEB-2.
RAEB with greater than 10% myeloblasts has a more aggres-
Refractory Cytopenia with Unilineage Dysplasia sive course, with a greater percentage of cases transforming
Presenting symptoms of RCUD are related to the cytopenia: to AML.53,57
namely, fatigue or shortness of breath if anemia is present,
increased infections from neutropenia, and petechiae, bruis- Myelodysplastic Syndrome with Isolated del(5q)
ing, or bleeding if thrombocytopenia is present. This category (5q Syndrome)
includes MDS cases with fewer than 1% blasts in the peripheral In patients who have only the deletion of 5q (5q), MDS rep-
blood and fewer than 5% blasts in the bone marrow. Dysplasia resents a fairly well-defined syndrome, affecting predominantly
must be present in more than 10% of a single myeloid lineage. women and occurring at a median age of 67. These patients
Included in RCUD is refractory anemia with only dyserythro- typically have refractory anemia without other cytopenias and/
poiesis (but fewer than 15% ringed sideroblasts), refractory or thrombocytosis, hypolobulated megakaryocytes, and ery-
neutropenia, and refractory thrombocytopenia.52 Although throid hypoplasia.58-60 There are fewer than 1% blasts in the
cytogenetic abnormalities may be seen in up to 50% of cases peripheral blood and Auer rods are not seen.61 Patients with
of refractory anemia, none are specific to the diagnosis. MDS with isolated del(5q) have long-term stable disease
Median survival is generally 2 to 5 years with only a 2% risk (median survival, 145 months). Recently the thalidomide ana-
of transformation to acute leukemia.52,53 logue lenalidomide has proven to be effective in patients with
isolated del(5q) as well as in those with del(5q) and additional
Refractory Anemia with Ringed Sideroblasts cytogenetic abnormalities.58-60
In RARS, anemia and dyserythropoiesis are present, and more
than 15% of the bone marrow erythroid precursors are ringed Myelodysplastic Syndrome, Unclassifiable
sideroblasts. To be considered a ringed sideroblast, an ery- The category of myelodysplastic syndrome, unclassifiable refers to
throid precursor must contain more than five iron granules per subtypes of MDS that initially lack the specific changes neces-
cell, and these iron-containing mitochondria must circle at sary for classification into other MDS categories. If characteris-
least one third of the nucleus (Figure 35-14).54,55 In the periph- tics of a specific subtype develop later, the case should be
eral blood there may be a dimorphic picture, with a population reclassified into the appropriate group.62
of hypochromic cells along with a majority of normochromic
cells. RARS occurs primarily in the older population. Mean Childhood Myelodysplastic Syndromes
survival is 69 to 108 months.56 Acquired sideroblastic anemia, De novo MDS in children is very rare, and although some of
which is not considered MDS, is discussed in Chapter 19. the characteristics of adult MDS are present, there are also some
distinct differences. The 2008 WHO classification introduced
Refractory Cytopenia with Multilineage Dysplasia a provisional category of refractory cytopenia of childhood. Several
RCMD is categorized by one or more cytopenias, dysplasia in authors have addressed this provisional category in detail.10,52,58
two or more myeloid cell lines, fewer than 1% blasts in periph-
eral blood, and fewer than 5% blasts in the bone marrow. In
MYELODYSPLASTIC/MYELOPROLIFERATIVE
RCMD, the myeloblasts do not contain Auer rods; if Auer rods
NEOPLASMS
are noted, the disorder is classified as RAEB-2.57 Some cases of
RCMD have more than 15% ringed sideroblasts, but the dys- The MDS/MPN category includes myeloid neoplasms with
poiesis in more than the erythroid line places them in the clinical, laboratory, and morphologic features that are charac-
RCMD, rather than the RARS, category.53 This distinction is teristic of both MDS and MPN. Included in this classification
important, because RCMD has a more aggressive course are chronic myelomonocytic leukemia, atypical chronic
than RARS.33 myeloid leukemia, juvenile myelomonocytic leukemia, and
540 PART V Leukocyte Disorders

Myelodysplastic/Myeloproliferative
BOX 35-5 Classification of Myelodysplastic Neoplasm, Unclassifiable
Syndromes/Myeloproliferative Neoplasms The designation MDS/MPN, unclassifiable is used for cases that
Chronic myelomonocytic leukemia meet the criteria for MDS/MPN but do not fit into one of the
Atypical chronic myeloid leukemia, BCR/ABL1 negative specified subcategories.69
Juvenile myelomonocytic leukemia
Myelodysplastic/myeloproliferative neoplasm, unclassifiable

From Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classification of CYTOGENETICS AND
tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
IARC Press.
MOLECULAR GENETICS
Cytogenetics
Chromosome abnormalities are found in about 50% of cases
MDS/MPN, unclassifiable, with a provisional subtype of refrac- of de novo MDS.70 Karyotype has a major effect on prognosis
tory anemia with ringed sideroblasts and thrombocytosis in MDS patients, and specific karyotypes can be used cautiously
(Box 35-5).52 to predict response to certain treatments.70 Balanced transloca-
tions, which are common among patients with AML, are found
Chronic Myelomonocytic Leukemia only rarely in cases of de novo MDS.19 Except for del(5q),
CMML is characterized by a persistent monocytosis of more no cytogenetic abnormality is specific to subtype. The most
than 1.0 monocytes 109/L, absence of the BCR/ABL1 fusion common abnormalities involve chromosomes 5, 7, 8, 11, 13,
gene, fewer than 20% blasts and promonocytes in the periph- and 20.9,19 The most common single abnormalities are trisomy
eral blood and bone marrow, and dysplasia in one or more 8 and monosomy 7.71,72 Less common abnormalities in MDS
myeloid cell line. Patients usually have an increased leukocyte are 12p, iso 17, 22, and loss of the Y chromosome.55,73
count with absolute monocytosis. Dysgranulopoiesis is evident,
but neutrophil precursors make up fewer than 10% of the total Molecular Alterations
leukocytes.63 Splenomegaly may be present due to infiltration Several molecular aberrations have been identified in MDS, but
of leukemic cells. Although cytogenetic abnormalities are these features have not been used in predicting prognosis,
found in up to 40% of patients, there are none specific for because previously such testing was not readily available.
CMML. Prognosis varies depending on the number of blasts Advances in molecular genetic testing have made testing more
plus promonocytes. If there are fewer than 5% blasts and available for routine use, and there is the potential that such
promonocytes in the peripheral blood and fewer than 10% information could be used to strengthen other prognostic indi-
in the bone marrow, the disease is classified as CMML-1 and cator schemes.74 Likewise, identification of genetic defects may
the prognosis is better than in those cases in which there allow development of targeted therapies.75 Although not spe-
are 5% to 19% blasts and promonocytes in the peripheral cific to MDS, the most common mutations include those in
blood or 10% to 19% in the bone marrow (classified as the TP53 gene74 and RUNX1/AML1.76,77 NRAS has been detected
CMML-2).52,63 in a small percentage of MDS patients.78

Atypical Chronic Myeloid Leukemia,


BCR/ABL1 Negative
PROGNOSIS
Atypical CML, BCR/ABL1 negative (aCML), is characterized by
leukocytosis with morphologically dysplastic neutrophils and Subtypes of MDS are divided into three risk groups based on
their precursors. Basophilia may be present, but is not a promi- morphologic subtype and length of survival and incidence of
nent feature. Multilineage dysplasia is common. The BCR/ABL1 transformation to AML. RCUD and RARS are considered low
fusion gene is not present, but a variety of other karyotypic risk. The intermediate-risk group includes RCMD and RAEB-1.
abnormalities may be seen. Dyspoiesis may be seen in all cell Cases of RAEB-2 comprise the high-risk group. Cytogenetic
lines, but is most remarkable in the neutrophils, which may features have been recognized as an important prognostic indi-
exhibit Pelger-Hutlike cells, hypogranularity, and bizarre cator in MDS, and the International Myelodysplastic Syndrome
segmentation.64,65 The prognosis is poor for patients with Working Group identified three major risk categories. Patients
aCML, who either progress to AML or succumb to bone marrow with a normal karyotype, isolated 5q, isolated 20q, and Y
failure.66 belong to the low-risk group. The high-risk group includes
patients with complex abnormalities, that is, three or more
Juvenile Myelomonocytic Leukemia abnormalities or an abnormality of chromosome 7. The
Juvenile myelomonocytic leukemia is a clonal disorder charac- intermediate-risk group includes those with all other abnor-
terized by proliferation of the granulocytic and monocytic cell malities.79 Three criteria (percentage of bone marrow blasts,
lines and affects children from 1 month to 14 years of age. cytopenias, and cytogenetic features) were combined to
There is a strong association with neurofibromatosis type 1.67 produce the International Prognostic Scoring System (IPSS) for
Allogeneic stem cell transplantation is effective in about 50% MDSs (Table 35-2). Median survival predicted by the IPSS is
of patients.68 as follows:
CHAPTER 35 Myelodysplastic Syndromes 541

TABLE 35-2 International Prognostic Scoring System for Myelodysplastic Syndromes


SCORE
Prognostic Variables 0 0.5 1 1.5 2
Percentage of bone marrow blasts <5% 5-10% 11-19% 20-30%*
Karyotype Good Intermediate Poor
Number of cytopenias 0-1 2-3

Adapted from Greenberg P, Cox E, LeBeau M, etal: International scoring system for evaluating prognosis in myelodysplastic syndromes, Blood 89:2079-2088, 1997.
Scores for risk groups are as follows:
Low: 0
Intermediate 1: 0.5-1
Intermediate 2: 1.5-2
High: 2.5
*This group is recognized as acute myeloid leukemia in the World Health Organization classification (see Chapter 36).

Karyotype categories are defined as follows:


Good: normal, Y, del(5q), del(20q) (as single abnormalities)
Poor: complex (3 abnormalities) or chromosome 7 abnormalities
Intermediate: other abnormalities

Cytopenias are defined as follows:


Hemoglobin <10 g/dL
Neutrophils <1.8 109/L
Platelets <100 109/L

Low: 5.7 years Lenalidomide (Revlimid; Celgene, Summit, N.J.), a thalido-


Intermediate 1: 3.5 years mide analogue that is less toxic than thalidomide, was approved
Intermediate 2: 1.2 years by the FDA in 2005 for use in patients with low- or intermediate-
High: 0.4 years risk MDS.85-87 It has shown remarkable promise, especially in
patients with the 5q chromosome arm deletion. Transfusion
independence was achieved in 64% of patients, and a median
TREATMENT
increase of 3.9g/dL of hemoglobin was achieved in patients
Treatment of MDS patients is challenging, because many are taking the drug. Complete cytogenetic remission was seen
older and have coexisting illnesses, and the heterogeneity of in 55% of MDS patients taking lenalidomide, whereas in
the disease makes use of one standard treatment impossible. MDS patients taking erythropoietin cytogenetic remission is
The overall goal of treatment is to provide improved quality of rare.85,86 Lenalidomide has immunomodulatory and antian
life and to prolong survival. giogenic effects.80,82 The apparent efficacy of lenalidomide
Supportive care has been the predominant mode of treat- must be weighed against its ability to cause significant
ment for most MDS patients, except those who qualify for stem myelosuppression.80,85
cell transplantation. Supportive care includes administration Ras is mutated in about 20% of MDS patients. Farnesyl-
of blood products (RBCs and platelets as necessary) and pre- transferase inhibitors interfere with this process.82,88 Patients
vention or treatment of infections with antibiotics. with high-risk MDS benefit from treatment with hypo
Recently, however, three drugs (lenalidomide, azacitidine, methylating agents such as azacitidine and, to a lesser extent,
and decitabine) have been approved by the U.S. Food and decitabine.80,89-91
Drug Administration (FDA) that show promise when used The only cure is hematopoietic stem cell transplantation.
either alone or in combination with other therapies.80 Thera- Patients with an IPSS score of intermediate 2 or higher and
pies are usually stratified into those used in low-risk disease patients with more than 10% blasts should be considered for
and those in higher-risk MDS cases. stem cell transplantation. Stem cell transplantation is most suc-
Treatment for patients with low-risk MDS is aimed at cessful in patients younger than age 70 with no comorbidity.9
maintaining residual function of the bone marrow through the
use of hematopoietic growth factors such as erythropoietin, Future Directions
thrombopoietin, and granulocyte colony-stimulating factor. As research addressing the role of apoptosis in MDS continues,
Although levels of these growth factors are often normal in future therapies may be aimed at controlling apoptosis, with
MDS patients, there is a subset of patients who respond to or without the use of chemotherapeutic agents. Because effec-
their use.80-82 tive treatment for MDS remains limited, it has been suggested
In patients with low-risk MDS, immunosuppressive therapy that patients be provided with information on the prognosis
with drugs such as antithymocyte globulin and cyclosporine for their type of MDS, available therapies, and success rates,
has resulted in decreased risk of leukemic tranformation.82-84 and take part in making decisions regarding their treatment.
Targeted molecular therapy is an option for patients whose As more is learned about the molecular biology of MDS, it
MDS does not respond to growth factor therapy. Some options may be possible to develop customized treatment plans for
include lenalidomide and farnesyltransferase inhibitors. individual patients.92
542 PART V Leukocyte Disorders

SUMMARY
MDSs are a group of clonal disorders characterized by progressive Other treatments that have met with limited success include che
cytopenias and dyspoiesis of the myeloid, erythroid, and mega motherapeutic agents and biologic response modifiers.
karyocytic cell lines. Currently, the only cure for MDS is bone marrow or hematopoietic
The dyspoiesis is evidenced by abnormal morphologic appearance stem cell transplantation.
and abnormal function of the cell lines affected. Future treatment possibilities include the use of apoptosis-
The WHO classification of MDSs is based on morphologic, mole controlling drugs.
cular, cytogenetic, and immunologic characteristics of blood cell
lines. Now that you have completed this chapter, go back and
Prognosis in MDS depends on several factors, including subtype, read again the case study at the beginning and respond
percentage of bone marrow blasts, cytopenias, and karyotypic to the questions presented.
abnormalities.
Treatment of MDS depends on the prognosis. If the prognosis is
favorable, patients may receive only supportive therapy.

R E V I E W Q UESTIONS
1. MDSs are most common in which age group? 6. A patient has anemia, oval macrocytes, and hyper
a. 2 to 10 years segmented neutrophils. Which of the following tests
b. 15 to 20 years would be most efficient in differential diagnosis of this
c. 25 to 40 years disorder?
d. Older than 50 years a. Serum iron and ferritin levels
b. Erythropoietin level
2. What is a major indication of MDS in the peripheral blood c. Vitamin B12 and folate levels
and bone marrow? d. Chromosome analysis
a. Dyspoiesis
b. Leukocytosis with left shift 7. A 60-year-old woman comes to the physician with fatigue
c. Normal bone marrow with abnormal peripheral and malaise. Her hemoglobin is 8g/dL, hematocrit is
blood features 25%, RBC count is 2.00 1012/L, platelet count is 550
d. Thrombocytosis 109/L, and WBC count is 3.8 109/L. Her WBC differen-
tial is unremarkable. Bone marrow shows erythroid hypo
3. An alert hematologist should recognize all of the following plasia and hypolobulated megakaryocytes; granulopoiesis
peripheral blood abnormalities as diagnostic clues in MDS appears normal. Ringed sideroblasts are rare. Chromo-
except: some analysis reveals the deletion of 5q only. Based on the
a. Oval macrocytes classification of this disorder, what therapy would be most
b. Target cells appropriate?
c. Agranular neutrophils a. Supportive therapy; lenalidomide if the disease
d. Circulating micromegakaryocytes progresses
b. Aggressive chemotherapy
4. For an erythroid precursor to be considered a ringed c. Bone marrow transplantation
sideroblast, the iron-laden mitochondria must encircle d. Low-dose cytosine arabinoside, accompanied by
how much of the nucleus? cis-retinoic acid
a. One quarter
b. One third 8. Which of the following is least likely to contribute to the
c. Two thirds death of patients with MDS?
d. Entire nucleus a. Neutropenia
b. Thrombocytopenia
5. According to the WHO classification of MDS, what per- c. Organ failure
centage of blasts would constitute transformation to an d. Neuropathy
acute leukemia?
a. 5%
b. 10%
c. 20%
d. 30%
CHAPTER 35 Myelodysplastic Syndromes 543

9. Into what other hematologic disease does MDS often 10. Chronic myelomonocytic leukemia is classified in the
convert? WHO system as:
a. Megaloblastic anemia a. A myeloproliferative neoplasm
b. Aplastic anemia b. Myelodysplastic syndrome, unclassified
c. AML c. MDS/MPN
d. Myeloproliferative disease d. Acute leukemia

REFERENCES
1. Layton DM, Mufti GJ: Myelodysplastic syndromes: their 18. Strom SS, Gu Y, Gruschkus SK, et al: Risk factors of
history, evolution and relation to acute myeloid leukemia. myelodysplastic syndromes: a case-control study. Leukemia
Blood 53:423-436, 1986. 19(11):1912-1918, 2005.
2. Mufti GJ, Galton DAG: Myelodysplastic syndromes: natural 19. Brunning RD, Orazi A, Germing U, et al: Myelodysplastic
history and features of prognostic significance. Clin Haematol syndromes/neoplasms, overview. In Swerdlow SH, Campo E,
15:953-971, 1986. Harris NL, et al, editors: WHO classification of tumours of hae
3. Cheson BD, Bennett JM, Kantarjian H, et al: Report of an matopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
international working group to standardize response criteria IARC Press, pp 88-93.
for myelodysplastic syndromes. Blood 96:3671-3674, 2000. 20. Mufti GJ: Pathobiology, classification, and diagnosis of
4. Bennett JM, Catovsky D, Daniel MT, et al: Proposals for the myelodysplastic syndrome. Best Pract Res Clin Haematol
classification of the myelodysplastic syndromes. Br J Haema 17(4):543-557, 2004.
tol 51:189-199, 1982. 21. Yoshida Y, Mufti GJ: Apoptosis and its significance in MDS:
5. Harris NL, Jaffe ES, Diebold J, et al: The World Health Orga- controversies revisited. Leuk Res 23:777-785, 1999.
nization classification of hematological malignancies report 22. Yoshida Y: Hypothesis: apoptosis may be the mechanism
of the Clinical Advisory Committee Meeting, Airlie House, responsible for the premature intramedullary cell death
Virginia, November 1997. Mod Pathol 13:193-207, 2000. in the myelodysplastic syndrome. Leukemia 7:144-146,
6. Vardiman JW, Harris NL, Brunning RD: The World Health 1993.
Organization (WHO) classification of the myeloid neo- 23. Yoshida Y, Stephenson J, Mufti GJ: Myelodysplastic syn-
plasms. Blood 100:2292-2302, 2002. dromes: from morphology to molecular biology: Part I. Clas-
7. Janssen JWG, Buschle M, Layton M, et al: Clonal analysis of sification, natural history and cell biology of myelodysplasia.
myelodysplastic syndromes: evidence of multipotential stem Int J Hematol 57:87-97, 1993.
cell origin. Blood 73:248-254, 1989. 24. Yoshida Y, Anzai N, Kawabata H: Apoptosis in myelodyspla-
8. Greenberg PL: Biologic nature of the myelodysplastic syn- sia: a paradox or paradigm. Leuk Res 19:887-891, 1995.
dromes. Acta Haematol 78(suppl 1):94-99, 1987. 25. DiGiuseppe JA, Kastan MB: Apoptosis in haematological
9. DeAngelo DJ, Stone RM: Myelodysplastic syndromes: biology malignancies. J Clin Pathol 50:361-364, 1997.
and treatment. In Hoffman R, Benz EJ Jr, Shattil SJ, 26. Lepelley P, Campergue L, Grardel N, et al: Is apoptosis a
et al, editors: Hematology: basic principles and practice, ed 5, massive process in myelodysplastic syndromes? Br J Haematol
Philadelphia, 2009, Churchill Livingstone Elsevier, chap 67. 95:368-371, 1996.
10. Hasle H, Niemeyer CM, Chessells JM, et al: A pediatric 27. Greenberg PL: Apoptosis and its role in the myelodysplastic
approach to the WHO classification of myelodysplastic and syndromes: implication for disease natural history and treat-
myeloproliferative diseases. Leukemia 17:277-282, 2003. ment. Leuk Res 22:1123-1126, 1998.
11. Niemeyer CM, Baumann I: Myelodysplastic syndrome in 28. Raza A, Gezer S, Mundle S, et al: Apoptosis in bone marrow
children and adolescents. Semin Hematol 45(1):60-70, 2008. biopsy samples involving stromal and hematopoietic cells in
12. Rollison DE, Howlader N, Smith MT, et al: Epidemiology of 50 patients with myelodysplastic syndromes. Blood 86:268-
myelodysplastic syndromes and chronic myeloproliferative 276, 1995.
disorders in the United States, 2001-2004, using data from 29. Rajapaksa R, Ginzton N, Rott LS, et al: Altered oncoprotein
the NAACCR and SEER programs. Blood 112:45-52, 2008. expression and apoptosis in myelodysplastic syndrome
13. Ma X, Does M, Raza A, et al: Myelodysplastic syndromes: marrow cells. Blood 88:4275-4287, 1996.
incidence and survival in the United States. Cancer 109:1536- 30. Parker JE, Mufti GJ: The role of apoptosis in the pathogenesis
1542, 2007. of the myelodysplastic syndromes. Int J Hematol 73:416-428,
14. Beris P: Primary clonal myelodysplastic syndromes. Semin 2001.
Hematol 26:216-233, 1989. 31. Wimazal F, Krauth M-T, Vales A, et al: Immunohistochemical
15. Tsukamoto N, Morita K, Maehara T, et al: Clonality in myelo- detection of vascular endothelial growth factor (VEGF) in the
dysplastic syndromes: demonstration of pluripotent stem cell bone marrow in patients with myelodysplastic syndromes:
origin using X-linked restriction fragment length polymor- correlation between VEGF expression and the FAB category.
phisms. Br J Haematol 83:589-594, 1993. Leuk Lymphoma 47:451-460, 2006.
16. Kroef MJ, Fibbe WE, Mout R, et al: Myeloid, but not lym- 32. Brunner B, Gunsilius E, Schumacher P, et al: Blood levels of
phoid cells carry the 5q deletion: polymerase chain reaction angiogenin and vascular endothelial growth factor are ele-
analysis of loss of heterozygosity using mini-repeat sequences vated in myelodysplastic syndromes and in some acute
on highly purified cell fractions. Blood 81:1849-1854, 1993. myeloid leukemia. J Hematother Stem Cell Res 11:119-125,
17. van Kamp H, Fibbe WE, Jansen RPM, et al: Clonal involve- 2002.
ment of granulocytes and monocytes, but not of T and B 33. Kjeldsberg CR, editor: Practical diagnosis of hematologic dis
lymphocytes and natural killer cells in patients with myelo- orders, ed 3, Chicago, 2000, ASCP Press, pp 369-397.
dysplasia: analysis by X-linked restriction fragment length 34. Tsurusawa M, Manabe A, Hayashi Y, et al: Therapy-related
polymorphism and polymerase chain reaction of phospho- myelodysplastic syndrome in childhood: a retrospective
glycerate kinase gene. Blood 80:1774-1980, 1992. study of 36 patients in Japan. Leuk Res 29:625-632, 2005.
544 PART V Leukocyte Disorders

35. Giles FJ, Koeffler HP: Secondary myelodysplastic syndromes 57. Brunning RD, Bennett JM, Matutes E, et al: Refractory cyto-
and leukemias. Curr Opin Hematol 1:256-260, 1994. penia with multilineage dysplasia. In Swerdlow SH, Campo
36. Rodak BF, Leclair SJ: The new WHO nomenclature: introduc- E, Harris NL, et al, editors: WHO classification of tumours of
tion and myeloid neoplasms. Clin Lab Sci 15:44-54, 2002. haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
37. Head DR, Kopecky K, Bennett JM, et al: Pathologic implica- IARC Press, pp 98-99.
tions of internuclear bridging in myelodysplastic syndrome. 58. Giagounidis AA, Germing U, Wainscoat, JS, et al: The 5q
An Eastern Cooperative Oncology Group/Southwest Oncol- syndrome. Hematology 9:271-277, 2004.
ogy Group Cooperative Study. Cancer 64:2199-2202, 1989. 59. Giagounidis AA, Germing U, Haase S, et al: Clinical, morpho-
38. Langenhuijsen MM: Neutrophils with ring-shaped nuclei in logical, cytogenetic, and prognostic features of patients with
myeloproliferative disease. Br J Haematol 58:227-230, 1984. myelodysplastic syndromes and del(5q) including band q31.
39. Doll DC, List AF: Myelodysplastic syndromes. West J Med Leukemia 18:113-119, 2004.
151:161-167, 1989. 60. Giagounidis AA, Haase S, Heinsch M, et al: Lenalidomide in
40. Seymour JF, Estey EH: The prognostic significance of Auer the context of complex karyotype or interrupted treatment:
rods in myelodysplasia. Br J Haematol 85:67-76, 1993. case reviews of del(5q) MDS patients with unexpected
41. Tricot G, De Wolf-Peeters R, Vlietinck R, et al: Bone marrow responses. Ann Hematol 86:133-137, 2007.
histology in myelodysplastic syndromes. Br J Haematol 61. Hasserjian RP, LeBeau MM, List AF, et al: Myelodysplastic
58:217-225, 1984. syndrome with isolated del(5q). In Swerdlow SH, Campo E,
42. Huff JD, Keung Y-K, Thakuri M, et al: Copper deficiency Harris NL, et al, editors: WHO classification of tumours of hae
causes reversible myelodysplasia. Am J Hematol 82(7):625- matopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
630, 2007. IARC Press, p 102.
43. Barbui T, Cortelazzo S, Viero P, et al: Infection and hemor- 62. Orazi A, Brunning RD, Baumann I: Myelodysplastic syn-
rhage in elderly acute myeloblastic leukemia and primary drome, unclassifiable. In Swerdlow SH, Campo E, Harris NL,
myelodysplasia. Hematol Oncol 11(suppl 1):15-18, 1993. et al, editors: WHO classification of tumours of haematopoietic
44. Mazzone A, Ricevuti G, Pasotti D, et al: The CD11/CD18 and lymphoid tissues, ed 4, Lyon, France, 2008, IARC Press,
granulocyte adhesion molecules in myelodysplastic syn- pp 103.
dromes. Br J Haematol 83:245-252, 1993. 63. Orazi A, Bennett JM, Germing U, et al: Chronic myelomono-
45. Mittelman M, Karcher D, Kammerman L, et al: High Ia (HLA- cytic leukemia. In Swerdlow SH, Campo E, Harris NL, et al,
DR) and low CD11b (Mo1) expression may predict early editors: WHO classification of tumours of haematopoietic and
conversion to leukemia in myelodysplastic syndromes. Am J lymphoid tissues, ed 4, Lyon, France, 2008, IARC Press,
Hematol 43:165-171, 1993. p 76.
46. Pomeroy C, Oken MM, Rydell RE, et al: Infection in the 64. Vardiman JW, Bennett JM, Bain BJ, et al: Atypical chronic
myelodysplastic syndromes. Am J Med 90:338-344, 1991. myeloid leukemia, BCR-ABL1 negative. In Swerdlow SH,
47. Boogaerts MA, Nelissen V, Roelant C, et al: Blood neutrophil Campo E, Harris NL, et al, editors: WHO classification of
function in primary myelodysplastic syndromes. Br J Haema tumours of haematopoietic and lymphoid tissues, ed 4, Lyon,
tol 55:217-227, 1983. France, 2008, IARC Press, pp 80-81.
48. Verhoef GE, Zachee P, Ferrant A, et al: Recombinant human 65. Xubo G, Xingguo L, Xiaanguo W, et al: The role of peripheral
erythropoietin for the treatment of anemia in the myelodys- blood, bone marrow aspirate and especially bone marrow
plastic syndromes: a clinical and erythrokinetic assessment. trephine biopsy in distinguishing atypical chronic myeloid
Ann Hematol 64:16-21, 1992. leukemia from chronic granulocytic leukemia and chronic
49. Merchav S, Nielsen OJ, Rosenbaum H, et al: In vitro studies myelomonocytic leukemia. Eur J Haematol 83:1292-1301,
of erythropoietin-dependent regulation of erythropoiesis in 2009.
myelodysplastic syndromes. Leukemia 4:771-774, 1990. 66. Breccia M, Biondo F, Latagliata R, et al: Identification of risk
50. Lintula R, Rasi V, Ikkala E, et al: Platelet function in preleu- factors in atypical chronic myeloid leukemia. Haematologica
kemia. Scand J Haematol 26:65-71, 1981. 91:1566-1568, 2006.
51. Raman BK, Van Slyck EJ, Riddle J, et al: Platelet function and 67. Niemeyer CM, Aric M, Basso G, et al: Chronic myelomono-
structure in myeloproliferative disease, myelodysplastic syn- cytic leukemia in childhood: a retrospective analysis of 110
drome and secondary thrombocytosis. Am J Clin Pathol cases. Blood 89:3534-3543, 1997.
91:647-655, 1989. 68. Locatelli F, Nllke P, Zecca M, et al: Hematopoietic stem cell
52. Vardiman JW, Thiele J, Arber DA, et al: The 2008 revision of transplantation (HSCT) in children with juvenile myelo-
the World Health Organization (WHO) classification of monocytic leukemia (JMML): results of the EWOG-MDS/
myeloid neoplasms and acute leukemia: rationale and EBMT trial. Blood 105:410-419, 2005.
important changes. Blood 114:937-951, 2009. 69. Vardiman JW, Bennett JM, Bain BJ, et al: Myelodysplastic/
53. Germing U, Strupp C, Kuemdgen A, et al: Prospective valida- myeloproliferative neoplasm, unclassifiable. In Swerdlow
tion of the WHO proposals for the classification of myelo- SH, Campo E, Harris NL, et al, editors: WHO classification of
dysplastic syndromes. Haematologica 91(12):1596-1604, tumours of haematopoietic and lymphoid tissues, ed 4, Lyon,
2006. France, 2008, IARC Press, pp 85-86.
54. Hast R: Sideroblasts in myelodysplasia: their nature and clini- 70. Haase D, Germing U, Schanz J, et al: New insights into the
cal significance. Scand J Haematol 36(suppl 45):53-55, 1986. prognostic impact of the karyotype in MDS and correlation
(abstract). with subtypes: evidence from a core dataset of 2124 patients.
55. Oscier DG: The myelodysplastic syndromes. In Hoffbrand AV, Blood 110;4385-4395, 2007.
Lewis SM, Tuddenham EGD, editors: Postgraduate 71. Bernasconi P, Klersy C, Boni M, et al: Incidence and
hematology, ed 4, Oxford, 1999, Butterworth-Heinemann, prognostic significance of karyotype abnormalities in de
pp 445-461. novo primary myelodysplastic syndromes: a study on 331
56. Hasserjian RP, Gattermann N, Bennett JM, et al: Refractory patients from a single institution. Leukemia 19:1424-1431,
anemia with ring sideroblasts. In Swerdlow SH, Campo E, 2005.
Harris NL, et al, editors: WHO classification of tumours of hae 72. Paulsson K, Johansson B: Trisomy 8 as the sole chromosomal
matopoietic and lymphoid tissues, ed 4, Lyon, France, 2008, aberration in acute myeloid leukemia and myelodysplastic
IARC Press, pp 96-97. syndromes. Pathol Biol (Paris) 55:37-48, 2007.
CHAPTER 35 Myelodysplastic Syndromes 545
73. Geddes AD, Bowen DT, Jacobs A: Clonal karyotype abnor- 83. Sloand EM, Wu CO, Greenberg P, et al: Factors affecting
malities and clinical progress in the myelodysplastic syn- response and survival in patients with myelodysplasia treated
drome. Br J Haematol 76:194-202, 1990. with immunosuppressive therapy. J Clin Oncol 26:2505-
74. Horiike S, Kita-Sasai Y, Nakao M, et al: Configuration of 2511, 2008.
the TP53 gene as an independent prognostic parameter of 84. Jonsova A, Neuwirtov R, Cermk J, et al: Cyclosporin A
myelodysplastic syndrome. Leuk Lymphoma 44:915-922, therapy in hypoplastic MDS patients and certain refractory
2003. anaemias without hypoplastic bone marrow. Br J Haematol
75. Cilloni D, Messa E, Messa F, et al: Genetic abnormalities as 100:304-309, 1998.
targets for molecular therapies in myelodysplastic syndromes. 85. List A, Kurtin S, Roe D, et al: Efficacy of lenalidomide in
Ann N Y Acad Sci 1089:411-423, 2006. myelodysplastic syndromes. N Engl J Med 352:549-557,
76. Chen CY, Lin LI, Tang JL, et al: RUNX1 gene mutation in 2005.
primary myelodysplastic syndromethe mutation can be 86. Moyer P: Lenalidomide reduces need for transfusions in
detected early at diagnosis or acquired during disease pro- patients with MDS. Available at: http://www.medscape.com/
gression and is associated with poor outcome. Br J Haematol viewarticle/505142. Accessed August 25, 2010.
139:405-414, 2007. 87. Witter D: Gaining momentum in MDS. Available at: http://
77. Harada H, Harada Y, Niimi H, et al: High incidence of www2.mdanderson.org/depts/oncolog/articles/06/7-8-julaug/
somatic mutations in the AML1/RUNX gene in myelodysplas- 7-8-06-1.html. Accessed August 25, 2010.
tic syndrome and low blast percentage myeloid leukemia 88. Lancet JE, Karp JE: Farnesyltransferase inhibitors in hemato-
with myelodysplasia. Blood 103:2316-2324, 2004. logic malignancies: new horizons in therapy. Blood 102:3880-
78. Epling-Burnette PK, List AF: Advancements in the molecular 3889, 2003.
pathogenesis of myelodysplastic syndrome. Curr Opin 89. Cheson BD, Bennett JM, Kantarijian H, et al: Report of an
Hematol 16:70-76, 2009. international working group to standardize response criteria
79. Greenberg P, Cox C, LeBeau MM: International scoring for myelodysplastic syndromes. Blood 96:3671-3674, 2000.
system for evaluating prognosis in myelodysplastic syn- 90. Silverman LR, Demakos EP, Peterson BL, et al: Randomized
dromes. Blood 89:2079-2088, 1997. controlled trial of azacytidine in patients with the myelodys-
80. Sekeres MA: Treatment of MDS; something old, something plastic syndrome: a study of the Cancer and Leukemia Group
new, something borrowed . Hematology Am Soc Hematol B. J Clin Oncol 20:2429-2440, 2002.
Educ Program 656-663, 2009. 91. Kornblith AB, Herndon JE 2nd, Silverman LR, et al: Impact
81. Hellstrm-Lindberg E, Malcovati L: Supportive care, growth of azacytidine on the quality of life of patients with myelo-
factors, and new therapies in myelodysplastic syndromes. dysplastic syndrome treated in a randomized phase trial III,
Blood Rev 22:75-91, 2008. a Cancer and Leukemia Group study B. J Clin Oncol 20:2441-
82. Loaiza-Bonilla A, Gore S, Carraway HE: Novel approaches for 2452, 2002.
myelodysplastic syndromes: beyond hypomethylating agents. 92. Scott BL, Deeg HJ: Myelodysplastic syndromes. Annu Rev Med
Curr Opin Hematol 17:104-109, 2010. 61:345-358, 2010.
36 Acute Leukemias
Susan J. Leclair and Bernadette F. Rodak

OUTLINE OBJECTIVES
Acute Lymphoblastic After completion of this chapter, the reader will be able to:
Leukemia
World Health Organization 1. Clinically distinguish between acute lymphoblastic and 4. Interpret the results of diagnostic tests for acute
Classification acute myeloid leukemias. leukemias.
Morphology 2. Characterize the diagnostic criteria used for these 5. Discuss tumor lysis syndrome, including risk, cause,
Immunophenotyping leukemias. and laboratory findings.
Prognosis 3. Compare and contrast acute lymphoblastic and 6. Discuss the rationale for the 2008 World Health
Genetics myeloid leukemias by morphology, presenting signs Organization classification of acute leukemias.
Types of Treatment and symptoms, laboratory findings, and prognosis.
Acute Myeloid Leukemia
Clinical Presentation
Subtypes of Acute
Myeloid Leukemia and
Related Precursor CASE STUDY
Neoplasms After studying the material in this chapter, the reader should be able to respond to the following
Acute Leukemias of case study:
Ambiguous Lineage
A 5-year-old child was seen by her family physician because of weakness and headaches. She had
been in good health except for the usual communicable diseases of childhood. Physical examina-
tion revealed a pale, listless child with multiple bruises. The WBC count was 15 109/L, the hemo-
globin was 8g/dL, and the platelet count was 90 109/L. She had abnormal cells in her peripheral
blood (Figure 36-1). Cytogenetic studies revealed hyperdiploidy.
1. What is the most likely diagnosis?
2. What characteristics of this disease indicate a positive prognosis?
3. What prognosis is associated with the hyperdiploidy?

Figure 36-1 Peripheral blood film for the patient in the case study (1000). (From Carr JH, Rodak BF: Clinical
hematology atlas, ed 3, St Louis, 2009, Saunders.)

546
CHAPTER 36 Acute Leukemias 547

U
nderstanding of the acute leukemias has undergone
a profound change. The paradigm shift from pheno- BOX 36-1 B Lymphoblastic Leukemia/Lymphoma
type assays (morphology and cytochemistry) to geno- with Recurrent Genetic Abnormalities (2008 World Health
type assays (karyotyping) to polymerase chain reaction and Organization Classification)
identification of single-nucleotide polymorphisms is clarifying B lymphoblastic leukemia/lymphoma with t(9;22)(q34;q11.2);BCR-ABL1
their cause and prognosis to an extent never before possible. B lymphoblastic leukemia/lymphoma with t(v;11q23);MLL rearranged
Although cell morphology will remain essential to the identi- B lymphoblastic leukemia/lymphoma with t(12;21)(p13;q22);TEL-AML1
fication of a leukemic process, molecular testing has begun to (ETV6-RUNX1)
dominate in the investigation of cause and therapy selection. B lymphoblastic leukemia/lymphoma with hyperdiploidy
According to the World Health Organization (WHO) clas- B lymphoblastic leukemia/lymphoma with hypodiploidy
sification, a finding of at least 20% blasts in the bone marrow B lymphoblastic leukemia/lymphoma with t(5;14)(q31;q32);IL3-IGH
is required for diagnosis of the majority of acute leukemias, B lymphoblastic leukemia/lymphoma with t(1;19)(q23;p13.3);E2A-PBX1
and testing must be performed for the presence or absence of (TCF3-PBX1)
genetic anomalies. The presence of certain genetic abnormali- From Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classification of
ties allows the diagnosis of a particular subtype of leukemia tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
regardless of blast count. Acute leukemia in individuals with a IARC Press.
history of malignancy with or without treatment is a distinct
category of its own. In-depth discussion of each of the subclas-
sifications is beyond the scope of this book; only the most
common entities are detailed here.

ACUTE LYMPHOBLASTIC LEUKEMIA


Acute lymphoblastic leukemia (ALL) is primarily a disease of
childhood and adolescence, accounting for 25% of all child-
hood cancers and up to 75% of childhood leukemias.1 Although
ALL is rare in adults, risk increases with age; most adult patients
are older than 65 years.2 Treatment of childhood ALL is a
triumph of modern hematology. A uniformly fatal disease
before 1970, good prognosis ALL in children is associated
with a 95% remission rate, and over 80% of patients are cured.1,3,4 Figure 36-2 Acute lymphoblastic leukemia. Large lymphoblast with
The subtype of ALL is an important prognostic indicator for prominent nucleoli and membrane irregularities (peripheral blood, 1000).
survival.5 Adults have a poorer outlook: 80% to 90% experi- (From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009,
ence complete remission, but the cure rate is less than 40%.6,7 Saunders.)
Only half of patients with ALL have leukocytosis, and many do
not have circulating lymphoblasts.8 Neutropenia, thrombocy- entities are linked with unique clinical, phenotypic, or prog-
topenia, and anemia are usually present, which leads to fatigue nostic features (Box 36-1). Cases of B-cell ALL (B-ALL) that do
(caused by anemia), fever (caused by neutropenia and infec- not exhibit the specific genetic abnormalities are classified as
tion), and mucocutaneous bleeding (caused by thrombocyto- B lymphoblastic leukemia/lymphoma, not otherwise specified.
penia). Lymphadenopathy, including enlargement, is often a Although 50% to 70% of patients with T lymphoblastic
symptom.2 Enlargement of the spleen (splenomegaly) and of leukemia/lymphoma have abnormal karyotypes, none of the
the liver (hepatomegaly) may be seen. Bone pain often results abnormalities is clearly associated with specific biologic fea-
from infiltration of leukemic cells into the bone covering tures, and thus T-ALL is not further subdivided.11
(periosteum).2 Eventual infiltration of malignant cells into the
meninges, testes, or ovaries occurs frequently, and lympho- Morphology
blasts can be found in the cerebrospinal fluid.9 Lymphoblasts vary in size but fall into two morphologic types.
In T-cell ALL (T-ALL), there may be a mass in the mediasti- The most common type seen is a small lymphoblast (1.0 to
num, the area in the central chest containing the heart and its 2.5 times the size of a normal lymphocyte) with scant blue
supporting nerves and vessels, thymus and other lymph nodes, cytoplasm and indistinct nucleoli (see Figure 36-1); the second
trachea, esophagus, and so forth.10 type of lymphoblast is larger (2 to 3 times the size of a lym-
phocyte) with prominent nucleoli and nuclear membrane
World Health Organization Classification irregularities10 (Figure 36-2). These cells may be confused with
The precursor lymphoid neoplasms are classified by the WHO the blasts of acute myeloid leukemia (AML).
into two major groups: B lymphoblastic leukemia/lymphoma
and T lymphoblastic leukemia/lymphoma.11 B lymphoblastic Immunophenotyping
leukemia/lymphoma is subdivided into seven subtypes that are Although morphology is the first tool used to distinguish ALL
associated with recurrent cytogenetic abnormalities. These from AML, immunophenotyping (see Chapter 33) and genetic
548 PART V Leukocyte Disorders

analysis (see Chapter 31) are the most reliable indicators of a mutation (Philadelphia chromosomepositive ALL), has the
cells origin. Because both B and T cells are derived from lym- worst prognosis among ALLs. It is more common in adults
phoid progenitors, both express CD34, terminal deoxynucleo- (25% of all ALL cases) than in children (2% to 4% of
tidyl transferase, and HLA-DR. Four types of ALL have been ALL cases). Imatinib, which has shown success in treating
identified by immunologic methods: early B-ALL (progenitor chronic myelogenous leukemia, has improved survival (see
B, pro-B, or pre-pre-B), intermediate (common) ALL, precursor Chapter 34).
B (pre-B) ALL, and T-ALL. B-ALL is characterized by specific B lymphoblastic leukemia/lymphoma with t(v;11q23);MLL
B-cell antigens that are expressed at different stages of B-cell rearranged is more common in very young infants, and the
development. In general B cells express CD19, CD20, CD22, translocation may even occur in utero.19 This leukemia has a
CD24, C79a, CD10, cytoplasmic , and PAX-5 (B-cell specific very poor prognosis. About 25% of childhood ALL cases show
activator protein). The degree of differentiation of B-lineage a t(12;21)(p13;q22);ETV6-RUNX1 translocation and appear to
lymphoblasts often correlates with genetics and plays an derive from a B-cell progenitor, rather than the hematopoietic
important role in treatment decisions.8,10,12 In the earliest stage stem cell.20 This translocation is rare in adults. In children, it
of differentiation (pre-pre B or pro-B), blasts express CD19, carries a very favorable prognosis, with a cure rate of over 90%.
cytoplasmic CD79a, cytoplasmic CD22, and nuclear TdT. The Hyperdiploidy in B lymphoblastic leukemia/lymphoma is
incidence of pro-B ALL is about 5% in children and 11% in common in childhood B-ALL, accounting for 25% of cases, but
adults. In intermediate B-ALL, also called common ALL, CD10 is much less common in adults. This genotype is associated
is expressed. CD10+ ALL is sometimes called common ALLa with a very favorable prognosis in children, but a poor prog-
(cALLa), because this group comprises 65% of childhood ALL nosis in adults.21 Conversely, hypodiploidy (less than 46 chro-
and 51% of adult ALL. In the most mature of the B-ALLs, called mosomes) conveys a poor prognosis in both children and
pre-B, cytoplasmic is present. Pre-B ALL accounts for 15% of adults. B lymphoblastic leukemia/lymphoma with t(5:14)
childhood cases and 10% of adult B-ALL.13 (q31;q32);IL3-IGH occurs in both children and adults, but
T-ALL is seen most often in teenaged males with a medias- is very rare, accounting for fewer than 1% of ALL cases.
tinal mass, elevated peripheral blast counts, meningeal involve- There are so few cases that the implication for prognosis is
ment, and infiltration of extra marrow sites.14 The common uncertain. B lymphoblastic leukemia/lymphoma with t(1;19)
T-cell markers CD2, CD4, CD5, and CD8 are usually present. (q23;p13.3);E2A-PBX1(TCF3-PBX1) accounts for about 6% of
In addition, cells may express CD3, CD1a, CD5, CD4, and B-ALL cases in children. It is less common in adult ALL. Newer
CD8. Although this immunophenotype classically has been therapies have improved the outcome.8
associated with a poor outcome, aggressive chemotherapy has Although an abnormal karyotype is found in 50% to 70%
resulted in improved prognosis; however, patients with T-ALL of cases of T-ALL/T lymphoblastic lymphoma, the abnormali-
are more likely to relapse sooner than patients with B-ALL, and ties are more random and none is associated with specific
CNS relapse is more common.15 T-ALL accounts for about 1% biologic features or outcome.
of childhood cases and 27% of adult cases.13
Types of Treatment
Prognosis At the morphologic and immunophenotypic levels, ALLs
Prognosis in ALL has improved dramatically over the past appear to be somewhat homogeneous, but at the genetic level,
decade as a result of improvement in algorithms for treat- ALLs comprise a heterogeneous group.7,10 Thus, a variety of
ment.10,16 The prognosis for ALL depends on age at the time of treatments are used. Patients first are classified into low-,
diagnosis, lymphoblast load (tumor burden), immunopheno- standard-, and high-risk groups so that treatment can be appro-
type, and genetic abnormalities. Children rather than infants priately directed. In general, children between the ages of 1 and
or teens do the best. Chromosomal translocations are the 10 years with a white blood cell (WBC) count of less than
strongest predictor of adverse treatment outcomes for children 50 109/L, no extramedullary disease, and rapid response to
and adults. Peripheral blood lymphoblast counts greater than initial therapy are classified into the low-risk group. High-risk
20 to 30 109/L, hepatosplenomegaly, and lymphadenopathy features include age younger than 1 year, extramedullary
all are associated with worse outcome. In addition, the pres- spread, and poor response to initial therapy. Genetic features
ence of an aberrant myeloid surface marker found in ALL (such as detailed previously are also considered. Adults, by virtue of
as CD66c) is associated with a poor outcome in adults.17 The their age, are considered to be in the high-risk group.2
effects of other variables previously associated with a poorer In general, treatment modalities include remission induc-
prognosis, such as sex and ethnic group, have been eliminated tion therapy, followed by consolidation (intensification), and
when patients have been given equal access to treatment in then continuation therapy to eliminate residual disease.
trials carried out at a single institution.18 Central nervous system therapy (intrathecal route) is often
included. Children with ALL may experience some paralysis or
Genetics ability regression as a result of intrathecal therapy, in which
Cytogenetic abnormalities are seen in the majority of B- and chemotherapeutic drugs are administered directly into the
T-cell ALL. In T-ALL there is less specificity and less correlation central nervous system via the cerebrospinal fluid.22 Methotrex-
with treatment outcome than in B-ALL. B lymphoblastic ate, commonly used to treat ALL, is a folate antagonist that
leukemia/lymphoma with the t(9;22)(q34;q11.2);BCR-ABL1 causes megaloblastosis on blood film or bone marrow smear
CHAPTER 36 Acute Leukemias 549

(see Chapter 20) and rapid severe leukocytopenia and throm- by hyperkalemia, hyperphosphatemia, hyperuricemia and
bocytopenia. Medications used for the treatment of adult ALL hyperuricosuria, and hypocalcemia.25,26 The hyperkalemia
include daunorubicin, vincristine, and prednisone. Because alone can be life threatening. Aggressive prophylactic measures
daunorubicin frequently causes cardiac complications, routine to prevent or reduce the clinical manifestations of tumor lysis
cardiac enzyme assays are required. Vincristine attacks micro- syndrome are critical.27,28
tubule formation, is toxic to the nervous system, and interferes
with platelet activity. Newer agents such as monoclonal anti- Subtypes of Acute Myeloid Leukemia and
bodies (e.g., anti-CD52) are assuming the role of lead therapy, Related Precursor Neoplasms
alone or in combination with more traditional chemothera- The 2001 WHO classification incorporated genetic abnormali-
peutic drugs.23 Peripheral blood stem cell transplantation is the ties into the classification of AML, and four categories of AML
last line of treatment for unresponsive or relapsed ALL. (See were identified. Since that time it has been recognized that not
Chapter 29 for more information on the treatment of WBC only genetic lesions recognizable at the microscopic level but
neoplasia.) also submicroscopic lesions cooperate to influence leukemo-
genesis, and seven subtypes of AML are included in the 2008
WHO classification.29 Laboratory diagnosis begins with a com-
ACUTE MYELOID LEUKEMIA
plete blood count, peripheral blood film examination, and
AML is the most common family of leukemias in children bone marrow aspirate and biopsy specimen examination. The
younger than 1 year of age, but is rare in older children and total WBC count is variable and may be normal, increased, or
adolescents. It is the most common type of leukemia in adults, decreased; anemia is usually present, along with significant
and the incidence increases with age. The French-American- thrombocytopenia. The bone marrow is usually hypercellular,
British (FAB) classification of AML was based on morphology and greater than 20% of cells typically are marrow blasts,
and cytochemistry; the WHO classification relies heavily on although if certain genetic abnormalities are present the 20%
cytogenetics and molecular characterization.8,11 blast threshold is not necessary for the diagnosis of AML.8 Each
category is discussed, and a summary of the classification is
Clinical Presentation presented in Box 36-2.
The clinical presentation of AML is nonspecific but reflects The 2008 WHO classification for myeloid malignancies has
decreased production of normal bone marrow elements. Most categorized AMLs with recurrent cytogenetic abnormalities
patients with AML have a total WBC count between 5 and into subgroups based on the primary cytogenetic aberrations
30 109/L, although the WBC count may range from 1 to (see Box 31-1).8,11
200 109/L. Myeloblasts are present in the peripheral blood
in 90% of patients. Anemia, thrombocytopenia, and neutrope- AML with Recurrent Genetic Abnormalities
nia give rise to the clinical findings of pallor, fatigue, fever with Acute Myeloid Leukemia with t(8;21)(q22;q22);RUNX1/
infections, bruising, and bleeding. In addition, disseminated RUNX1T1. The t(8;21)(q22;q22);RUNX1/RUNX1T1 muta-
intravascular coagulation and other bleeding abnormalities tion is found in about 5% of AML cases. Seen predominantly
can be significant.24 Infiltration of malignant cells into the in children and young adults, AML with this translocation has
gums and other mucosal sites and skin also can be seen. myeloblasts with dysplastic granular cytoplasm, Auer rods, and
Bone and joint pain are the first symptoms in only 25% some maturation (Figure 36-3), similar to the FAB M2 classifi-
of patients, unlike in ALL, in which such pain is common.25 cation (see later in chapter). Various anomalies, such as
Splenomegaly is seen in half of AML patients, but lymph pseudoPelger-Hut cells and hypogranulation, can be seen.
node enlargement is rare. Cerebrospinal fluid involvement Eosinophilia is possible.30 The diagnosis of this subtype is
in AML is rare and does not seem to be as ominous a sign as based on the genetic abnormality, regardless of blast count.8
in ALL. Patients with AML tend to have few symptoms related
to the central nervous system, even when it is infiltrated
by blasts.
Common abnormalities in laboratory test results include BOX 36-2 Acute Myeloid Leukemia and Related
hyperuricemia (caused by increased cellular turnover), hyper- Precursor Neoplasms (2008 World Health Organization
phosphatemia (due to cell lysis), and hypocalcemia (the latter Classification)
two are also involved in progressive bone destruction). Hypo- Acute myeloid leukemia with recurrent genetic abnormalities
kalemia is also common at presentation. During induction Acute myeloid leukemia with myelodysplasia-related changes
chemotherapy, especially when the WBC is quite elevated, Therapy-related myeloid neoplasms
tumor lysis syndrome may occur. Tumor lysis syndrome is a Acute myeloid leukemia, not otherwise specified
group of metabolic complications that can occur in patients Myeloid sarcoma
with malignancy, most notably lymphomas and leukemias, Myeloid proliferations related to Down syndrome
with and without treatment of the malignancy.26 These Blastic plasmacytoid dendritic cell neoplasm
complications are caused by the breakdown products of From Swerdlow SH, Campo E, Harris NL, etal, editors: WHO classification of
dying cancer cells, which in turn cause acute uric acid nephrop- tumours of haematopoietic and lymphoid tissues, ed 4, Lyon, France, 2008,
athy and renal failure. Tumor lysis syndrome is characterized IARC Press.
550 PART V Leukocyte Disorders

Figure 36-3 Acute myeloid leukemia with t(8;21). Myeloblasts with


granular cytoplasm and some maturation (bone marrow, 500). (From Carr
JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009, Saunders.)

B
Figure 36-5 Acute myeloid leukemia with t(15;17), or promyelocytic
leukemia. A, Low-power view of the more common hypergranular variant
(peripheral blood, 500). B, Oil immersion view of the microgranular variant
showing bilobed nuclear features (peripheral blood, 1000). (B from Carr
JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009, Saunders.)

Figure 36-4 Acute myeloid leukemia with inv(16). There is an increase


for relapse.8,30 The remission rate is good, but only one half of
in myeloid and monocytic lines. Eosinophilia may also be present (peripheral
blood, 1000). (From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, patients are cured.31
St Louis, 2009, Saunders.)
Acute Myeloid Leukemia with t(15;17)(q22;q12);PML-
RARA. Also known as acute promyelocytic leukemia (APL),
Prognosis is generally favorable, but may be negatively impacted AML with the t(15;17)(q22;q12);PML-RARA mutation com-
if unfavorable additional abnormalities, such as monosomy prises 5% to 10% of AML cases. It occurs in all age groups but
7, occur.30 is seen most commonly in young adults. This disorder is char-
acterized by a differentiation block at the promyelocytic stage.
Acute Myeloid Leukemia with inv(16)(p13.1q22) or The abnormal promyelocytes are considered to be comparable
t(16;16)(p13.1;q22);CBFB-MYH11. Accounting for appro to blasts for the purpose of diagnosis. Detection of the 15;17
ximately 5% to 8% of all AML cases, core-binding factor (CBF) translocation is sufficient for diagnosis regardless of blast
AML occurs at all ages, but is found predominantly in younger count.8,30 Characteristic of this presentation are the abnormal
patients.8 Cytogenetic hallmarks of CBF AML may represent hypergranular promyelocytes, some with Auer rods (Figure
cooperative events in CBF AML leukemogenesis and include 36-5). When Auer rod bundles are found, the cell is called a
mutations in the KIT, FLT3, JAK2, and RAS genes. The genetic faggot cell. When promyelocytes release primary granule con-
aberration is sufficient for diagnosis regardless of blast count.8,31 tents, their procoagulant activity initiates disseminated intra-
Myeloblasts, monoblasts, and promyelocytes are seen in the vascular coagulation; however, thromboembolic events may
peripheral blood and bone marrow. In the bone marrow there occur at presentation and during treatment.32 In one variant of
may be eosinophilia with dysplastic changes (Figure 36-4). The APL, the granules are so small that, because of the limits of
incidence of extramedullary disease is higher than in most light microscopy, the cells give the appearance of having no
types of AML, and the central nervous system is a common site granules. This microgranular variant, accounting for 30% to
CHAPTER 36 Acute Leukemias 551

Acute Myeloid Leukemia with inv(3)(q21q26.2) or


t(3;3)(q21;q26.2);RPN1-EVI1. AML with inv(3)(q21q26.2)
or t(3;3)(q21;q26.2);RPN1-EVI1 occurs primarily in adults and
represents about 1% to 2% of adult AML cases. Patients gener-
ally present with anemia and a normal platelet count, although
thrombocytopenia or thrombocytosis (with or without previ-
ous myelodysplastic syndrome [MDS]) may be present. Hepa-
tosplenomegaly is fairly common. Blast morphology is variable.
Dyspoiesis is evidenced by hypogranular neutrophils, pseudo
Pelger-Hut anomaly, and giant and bizarre platelets, and bare
megakaryocytic nuclei may be present. Red cell morphologic
abnormalities are generally absent to mild. In the bone marrow
there may be increased and dysplastic megakaryoblasts. Prog-
nosis in this aggressive disorder is bleak.8,30
Figure 36-6 Acute myeloid leukemia with t(9;11) abnormalities. Both
monoblasts and immature monocytes are increased. (bone marrow, 500). Acute Myeloid Leukemia (Megakaryoblastic) with
t(1;22)(p13;q13);RBM15-MKL1. AML with the t(1;22)
(p13;q13);RBM15-MKL1 mutation represents fewer than 1%
40% of APL cases, may be confused with other presentations of AMLs. It occurs primarily in patients with Down syndrome.
of AML, but the presence of occasional Auer rods, the but- Age at presentation is younger than 3 years, with most
terfly or bowtie nucleus, and the clinical presentation are cases occurring in infants younger than 6 months. There is
clues. Treatment must include a resolution of the disseminated marked organomegaly, anemia, and thrombocytopenia. WBC
intravascular coagulation. The genetic abnormality seen in this counts are moderately elevated. Blasts have typical features
condition results in the ability of the cell to ingest vitamin A, of megakaryoblasts (see Chapter 13); that is, they are 12 to
and a successful treatment is to initiate maturation with a 18m with fine reticular chromatin and a round or slightly
vitamin A modification, all-trans-retinoic acid (ATRA), and indented nucleus. There may be cytoplasmic budding, and
arsenic trioxide.33 In adults who achieve a complete remission micromegakaryocytes may be present. Marrow fibrosis may
the prognosis is better than for any other type of AML.30 There cause a dry tap or aparticulate aspirate.8,30 Prognosis, once
are a few variants in RARA translocations that confer a poor considered poor, has improved with the use of intensive AML
diagnosis because the cells do not respond to ATRA therapy.8,11 chemotherapy.34

Acute Myeloid Leukemia with t(9;11)(p22;q23);MLLT3- Acute Myeloid Leukemia with


MLL. AML with t(9;11)(p22;q23);MLLT3-MLL represents a Myelodysplasia-Related Changes
specific subgroup of the previous classification of AML with The subcategory of AML with myelodysplasia-related changes
11q23 abnormalities, and AMLs with other MLL abnormalities incorporates leukemias with at least 20% blasts, multilineage
should not be placed in this group.11 AML with t(9;11) is a rare dysplasia, a history of MDS or MDS/myeloproliferative
leukemia (6% of AML cases) that presents with an increase neoplasm (MPN); or a specific MDS-associated cytogenetic
in monoblasts and immature monocytes (Figure 36-6). The abnormality and the absence of AML with recurrent genetic
blasts are large with abundant cytoplasm and fine nuclear chro- abnormalities.30,35 Significant dysplastic morphology includes
matin. The cells may have motility, with pseudopodia seen pancytopenia with neutrophil hypogranulation or hypergranu-
frequently. Granules and vacuoles can be observed in the lation, pseudoPelger-Hut cells, and unusually segmented
blasts. Typically this disease occurs in children and may be nuclei. Erythrocyte precursors have vacuoles, karyorrhexis,
associated with gingival and skin involvement and/or dissemi- megaloblastoid features, and ringed sideroblasts. There may
nated intravascular coagulation. The prognosis is intermediate be dysplastic micromegakaryocytes and dysplastic megakaryo-
to poor.30 cytes. Genetic findings are similar to those found in MDS, with
complex karyotypes and 7/del(7q) and 5/del(5q) being the
Acute Myeloid Leukemia with t(6;9)(p23;q34);DEK- most common8,36 (see Chapter 35). AML with myelodysplasia
NUP214. The rare subtype of AML with the t(6;9)(p23;q34); affects primarily older adults and has a poor prognosis.30,35
DEK-NUP214 mutation comprises only 1% of AML cases. It
occurs in both children and adults at a median age of 13 in Therapy-Related Myeloid Neoplasms
children and 35 in adults.8 There is often pancytopenia. Blast Treatment with alkylating agents, radiation, or topoisomerase
morphology is variable, but cells may have monocytic features II inhibitors has been associated with the development of a
or Auer rods. There is dyspoiesis in all cell lines; about 50% of secondary AML, MDS, or MDS/MPN.8,30,37,38 These therapy-
cases demonstrate mild basophilia. In this subtype the pres- related neoplasms account for 10% to 20% of AMLs, MDSs,
ence of at least 20% blasts is required for diagnosis. Prognosis and MDSs/MPNs. Generally these disorders occur following
is generally poor, and patients may benefit from allogeneic treatment for a prior malignancy, but have also been associated
stem cell transplantation.8,30 with intensive treatment of patients with nonmalignant
552 PART V Leukocyte Disorders

TABLE 36-1 French-American-British Classification of


the Acute Myeloid Leukemias
Subtype Description
M0 Acute myeloid leukemia, minimally differentiated
M1 Acute myeloid leukemia without maturation
M2 Acute myeloid leukemia with maturation
M3 Acute promyelocytic leukemia
M4 Acute myelomonocytic leukemia
M4eo Acute myelomonocytic leukemia with eosinophilia
M5a Acute monocytic leukemia, poorly differentiated
M5b Acute monocytic leukemia, well differentiated
M6 Acute erythroleukemia
M7 Acute megakaryocytic leukemia

Data from Bennett JM, Catovsky D, Daniel MT, etal: Proposals for the classification
of the acute leukemias. French-American-British (FAB) co-operative group, Br J Hae- Figure 36-7 Acute myeloid leukemia, minimally differentiated (French-
matol 33:451-458, 1976; and Bennett JM, Catovsky D, Daniel MT, etal: Proposed American-British classification M0). Blasts lack myeloid morphologic features
revised criteria for the classification of acute myeloid leukemia. A report of the French-
and yield negative results with myeloperoxidase and Sudan black B staining.
American-British Cooperative Group, Ann Intern Med 103:620-625, 1985.
Auer rods are not seen. CD34 is frequently present (bone marrow, 500).
(From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009,
Saunders.)
disorders requiring cytotoxic therapy.8,37,39 Therapy-related
myeloid neoplasms are similar in morphology to AML with
myelodysplasia, monocytic/monoblastic leukemia, or AML
with maturation, and the prognosis is generally poor, although
therapy-related neoplasms with the t(15;17) and inv(16)
mutations behave more like the de novo counterparts.8,30

Acute Myeloid Leukemia, Not Otherwise Specified


Because the leukemias in the not otherwise specified category
do not fit easily into the WHO subtypes described earlier, they
are grouped according to morphology, flow cytometric pheno-
typing, and limited cytochemical reactions, as in the FAB clas-
sification. The FAB classification was based on the cell of origin,
degree of maturity, cytochemical reactions, and limited cytoge-
netic features (Table 36-1).40,41 A blast percentage of at least
20% in the peripheral blood or bone marrow is required for
diagnosis. This category accounts for about 25% of all AML, Figure 36-8 Acute myeloid leukemia without maturation (French-
but as more genetic subgroups are recognized, the number in American-British classification M1). Blasts constitute 90% of the nonery-
this group will diminish.11 throid cells; there is less than 10% maturation of the granulocytic series
beyond the promyelocyte stage (bone marrow, 500).
Acute Myeloid Leukemia with Minimal Differentiation.
The blasts in AML with minimal differentiation are CD13+,
CD33+, CD34+, and CD117+ (Figure 36-7).8,42 Auer rods typi- Auer rods and usually give positive results with myeloperoxi-
cally are absent, and there is no clear evidence of cellular matu- dase or Sudan black B stains.8,30
ration. The cells yield negative results with the cytochemical
stains myeloperoxidase and Sudan black B. These cases account Acute Myeloid Leukemia with Maturation. AML with
for less than 5% of AML, and patients are generally either maturation is a common variant that presents with greater
infants or older adults. than 20% blasts, at least 10% maturing cells of neutrophil
lineage (Figure 36-9), and fewer than 20% precursors with
Acute Myeloid Leukemia without Maturation. Closely monocytic lineage. Auer rods and other aspects of dysplasia are
aligned with the blasts in minimally differentiated AML, the present.8
blasts in AML without maturation are also CD13+, CD33+, and
CD117+, and CD34 is present in about 70% of cases (Figure Acute Myelomonocytic Leukemia. One of the more
36-8).8 Blasts may comprise 90% of nonerythroid cells in the flamboyant AML presentations is characterized by a signifi-
bone marrow, and fewer than 10% of the leukocytes show cantly elevated WBC count and the presence of myeloid and
maturation to the promyelocyte stage or beyond. Blasts have monocytoid cells in the peripheral blood and bone marrow
CHAPTER 36 Acute Leukemias 553

Figure 36-9 Acute myeloid leukemia with maturation. Blasts constitute


20% or more of the nucleated cells of the bone marrow, and there is matura-
tion beyond the promyelocyte stage in more than 10% of the nonerythroid
cells (bone marrow, 1000).

B
Figure 36-11 A, Acute monoblastic leukemia. More than 80% of the
bone marrow cells are of monocytic origin (bone marrow, 500). B, Acute
monocytic leukemia with promonocytes. Promonocytes are considered blast
equivalents. (B from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
St Louis, 2009, Saunders.)
Figure 36-10 Acute myelomonocytic leukemia. Both myeloid and mono-
cytic cells are present. Monocytic cells comprise at least 20% of all marrow
cells, with monoblasts and promonocytes present (peripheral blood, 1000). prominent nucleoli (Figure 36-11, A). When some evidence of
(From Carr JH, Rodak BF: Clinical hematology atlas, ed 3, St Louis, 2009, maturation is present, the cells are called promonocytes. Pro-
Saunders.) monocytes in monocytic leukemias with differentiation are
considered to be blast equivalents (see Figure 36-11, B). Non-
(Figure 36-10). Monocytic cells (monoblasts and promono- specific esterase testing usually yields positive results. Acute
cytes) constitute at least 20% of all marrow cells. The mono- monoblastic/monocytic leukemia comprises fewer than 5% of
blasts are large with abundant cytoplasm containing small cases of AML and is most common in younger individuals.
granules and pseudopodia. The nucleus is large and immature Extramedullary involvement, including cutaneous and gingival
and may contain multiple nucleoli. Promonocytes also are infiltration, and bleeding disorders are common. Nonspecific
present and may have contorted nuclei. The cells are positive cytogenetic abnormalities are seen in most cases.8,43
for the myeloid antigens CD13 and CD33 and the monocytic
antigens CD14, CD4, CD11b, CD11c, CD64, and CD36. Non- Acute Erythroid Leukemia. According to the WHO
specific cytogenetic changes are found in most cases.8 classification, there are two subtypes of acute erythroid leuke-
mia, based on the presence of a significant component of
Acute Monoblastic and Monocytic Leukemias. In myeloblasts. The first is acute erythroleukemia (erythroid/
these leukemias, divided into monoblastic and monocytic myeloid), in which 50% or more of nucleated bone marrow
based on the degree of maturity of the monocytic cells present cells are normoblasts, and greater than 20% are myeloblasts.
in the marrow and peripheral blood, more than 80% of the In the FAB classification, this subtype was known as M6.
marrow cells are of monocytic origin. These cells are CD14+, The second type is pure erythroid leukemia. In this type 50%
CD4+, CD11b+, CD11c+, CD36+, CD64+, and CD68+. Blasts are or more nucleated cells are pronormoblasts and 30% or more
large with abundant, often agranular cytoplasm and large are basophilic normoblasts. Together these two erythroid
554 PART V Leukocyte Disorders

components comprise more than 80% of the bone marrow. Acute Megakaryocytic Leukemia. Patients with acute
The myeloblast component is not significant. Complex megakaryocytic leukemia usually have cytopenias, although
rearrangements and hypodiploid chromosome number are some may have thrombocytosis. Dysplastic features are often
common. Chromosomes 5 and 7 are frequently affected.8 present in all cell lines. Diagnosis requires the presence of at
The red blood cell (RBC) precursors have significant dys- least 20% blasts, of which at least 50% must be of megakaryo-
plastic features, such as multinucleation, megaloblastoid asyn- cyte origin. This category excludes AML with MDS-related
chrony, and vacuolization. Periodic acidSchiff staining yields changes and Down-related cases, as well as those with recurrent
positive results in the normoblasts, which stain either in a genetic abnormalities as discussed previously.
diffuse (erythroid/myeloid) or block (pure erythroid) pattern. Megakaryoblast diameters vary from that of a small lym-
The nucleated RBCs in the peripheral blood may account for phocyte to three times their size. Chromatin is delicate with
more than 50% of the total number of nucleated cells. Ringed prominent nucleoli. Immature megakaryocytes may have light
sideroblasts, Howell-Jolly bodies, and other inclusions may be blue cytoplasmic blebs (Figure 36-13, A). Megakaryoblasts are
present (Figure 36-12). Abnormal megakaryocytes may be identified by immunostaining, employing antibodies specific
seen. Both types of erythroid leukemia have an aggressive and for cytoplasmic von Willebrand factor or platelet membrane
rapid clinical course.8 antigens CD41 (glycoprotein IIb), CD42b (glycoprotein Ib)
(Figure 36-13, B), or CD61 (glycoprotein IIIa).

Myeloid Sarcoma
Myeloid sarcoma refers to extramedullary proliferation of blasts
of one or more myeloid lineages that disrupts tissue architec-
ture. Tissue architecture must be effaced for the neoplasm to
qualify for this diagnosis.8,30

Myeloid Proliferations Related to Down Syndrome


Unique patterns of malignancy occur in persons with trisomy
21 resulting in Down syndrome. Somatic mutations of the
GATA1 gene have also been detected and are linked to both
leukemogenesis and high cure rates.44 Approximately 10% of
newborns with Down syndrome present with transient abnor-
Figure 36-12 Acute erythroid leukemia. Erythroid precursors showing mal myelopoiesis, which is morphologically indistinguishable
dysplastic features, including multinucleation and megaloblastic asynchrony from AML. Spontaneous remission generally occurs within a
(bone marrow, 500). (From Carr JH, Rodak BF: Clinical hematology atlas, few months.8,44 Among individuals with Down syndrome
ed 3, St Louis, 2009, Saunders.) there is a fiftyfold increased incidence of AML during the first

A B
Figure 36-13 Acute megakaryocytic leukemia. A, Note heterogeneity of blasts, one small with scant cytoplasm, two with cytoplasmic blebbing, and one
quite large (peripheral blood 1000). B, Positive reaction for CD42b (bone marrow, 1000). (A from Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
St Louis, 2009, Saunders.)
CHAPTER 36 Acute Leukemias 555

5 years of life compared with individuals without Down syn-


ACUTE LEUKEMIAS OF
drome. The leukemia is of megakaryocytic lineage and young
AMBIGUOUS LINEAGE
children respond well to chemotherapy, although older chil-
dren do not fare as well.8,44 Acute leukemias of ambiguous lineage include leukemia in
which there is no clear evidence of differentiation along a
single cell line. In some cases, there are no lineage-specific
Blastic Plasmacytic Cell Neoplasm antigens, whereas in others antigens of more than one lineage
Blastic plasmacytic cell neoplasm is a rare clinically aggressive are expressed to such a degree that it is not possible to deter-
tumor derived from precursors of plasmacytoid dendritic cells. mine a specific lineage. The 2008 WHO classification signifi-
It presents with skin lesions and may ultimately progress to cantly revised the criteria for this designation, and the interested
involve peripheral blood and bone marrow.8,11 reader is referred to that classification.8

SUMMARY
Only half of patients with ALL have leukocytosis and many do not Although morphology is the first tool in distinguishing ALL from
have circulating lymphoblasts, but neutropenia, thrombocytope- AML, immunophenotyping is often the only reliable indicator of a
nia, and anemia are usually present. cells origin.
In children ALL is a disease in which the good prognosis sub- AML is the most common leukemia in children younger than 1
types are associated with a 95% rate of complete remission, but year of age. It is rare in older children and adolescents. In adults,
adults with ALL have a poorer outlook. its incidence increases with age.
Infiltration of malignant cells into the meninges can occur, with The clinical presentation of a patient with AML is nonspecific and
lymphoblasts found in the cerebrospinal fluid, testes, and ovaries. reflects the decreased production of normal bone marrow ele-
Prognosis in ALL depends primarily on age at the time of diagno- ments, an elevated WBC count, and the presence of myeloblasts.
sis, lymphoblast load (tumor burden), and immunophenotype. Anemia, thrombocytopenia, and neutropenia give rise to the clini-
Chromosomal translocations seem to be the strongest predictor of cal findings of pallor, fatigue, bruising and bleeding, and fever with
adverse treatment outcomes for children and adults. infections.
The t(12;21) marker is found in a significant number of patients The classification of AML is complicated by the presence or absence
with childhood ALL. of multiple cell lines defined as myeloid in origin, specific cells
The presence of the Philadelphia chromosome, t(9;22), in ALL is within these cell lines, and specific karyotype abnormalities.
an indicator of a significantly worse prognosis. Leukemias with ambiguous lineage include leukemias in which
There are two main subtypes of ALL according to the WHO clas- there is no clear evidence of differentiation along a single cell line.
sification system: B lymphoblastic leukemia/lymphoma and T
lymphoblastic leukemia/lymphoma. Now that you have completed this chapter, go back and
Tumor lysis syndrome is an increasingly common complication of read again the case study at the beginning and respond
treatment, especially in patients with a high tumor burden. to the questions presented.

R E V I E W Q UESTIONS
1. According to the WHO classification, except in leukemias 3. Which of the following would be considered a sign of
with specific genetic anomalies, the minimal percentage of potentially favorable prognosis in children with ALL?
blasts necessary for a diagnosis of acute leukemia is: a. Hyperdiploidy
a. 10% b. Presence of CD 19 and 20
b. 20% c. Absence of trisomy 8
c. 30% d. Presence of BCR/ABL gene
d. 50%
4. Signs and symptoms of cerebral infiltration with blasts are
2. A 20-year-old patient has an elevated WBC count with 70% more commonly seen in:
blasts, 4% neutrophils, 5% lymphocytes, and 21% mono- a. AML with recurrent cytogenetic abnormalities
cytes in the peripheral blood. Eosinophils with dysplastic b. Therapy-related myeloid neoplasms
changes are seen in the bone marrow. AML with which of c. AML with myelodysplasia-related changes
the following karyotypes would be most likely to be seen? d. ALL
a. AML with t(8;21)(q22:q22)
b. AML with t(16;16)(p13;q22)
c. AML with t(15;17)(q22;q12)
d. AML with t(9;11)(p22;q23)
556 PART V Leukocyte Disorders

5. An oncology patient exhibiting signs of renal failure 8. A 20-year-old patient presents with fatigue, pallor, easy
with seizures after initial chemotherapy may potentially bruising, and swollen gums. Bone marrow examination
develop: reveals 82% cells with delicate chromatin and prominent
a. Hyperleukocytosis nucleoli that are CD14+, CD4+, CD11b+, and CD36+.
b. Tumor lysis syndrome Which of the following acute leukemias is likely?
c. Acute leukemia secondary to chemotherapy a. Minimally differentiated leukemia
d. Myelodysplasia b. Leukemia of ambiguous lineage
c. Acute monoblastic/monocytic leukemia
6. Disseminated intravascular coagulation is more often seen d. Acute megakaryoblastic leukemia
in association with leukemia characterized by which of the
following mutations? 9. Pure erythroid leukemia is a disorder involving:
a. t(12;21)(p13;q22) a. Pronormoblasts only
b. t(9;22(q34;q11.2) b. Pronormoblasts and basophilic normoblasts
c. inv(16)(p13q22) c. All forms of developing RBC precursors
d. t(15;17)(q22;q12) d. Equal numbers of pronormoblasts and myeloblasts

7. Which of the following leukemias affects primarily chil- 10. A patient with normal chromosomes has a WBC count of
dren, is characterized by an increase in monoblasts and 3.0 109/L and dysplasia in all cell lines. There are 60%
monocytes, and often is associated with gingival and skin blasts of varying sizes. The blasts stain positive for CD61.
involvement? The most likely type of leukemia is:
a. Pre-B lymphoblastic leukemia a. Acute lymphoblastic
b. Pure erythroid leukemia b. Acute megakaryoblastic
c. AML with t(9;11)(p22;q23) c. Acute monoblastic
d. AML with t(15;17((q22;q12) d. AML with t(15;17)

REFERENCES
1. Stanulla M, Schrauder A: Bridging the gap between the north 11. Vardiman JW, Thiele J, Arber DA, et al: The 2008 revision of
and south of the world: the case of treatment response in the World Health Organization (WHO) classification of
childhood acute lymphoblastic leukemia. Haematologica myeloid neoplasms and acute leukemia: rationale and
94:748-752, 2009. important changes. Blood 114:937-951, 2009.
2. Redaelli A, Laskin BL, Stephens JM: A systematic literature 12. Thomas DA, OBrien S, Kantajian HM: Monoclonal antibody
review of the clinical and epidemiological burden of acute therapy with rituximab for acute lymphoblastic leukemia.
lymphoblastic leukemia (ALL). Eur J Cancer Care (Engl) Hematol Oncol Clin North Am 23:949-971, 2009.
14:53-62, 2005. 13. Hoelzer D, Gkbuget N: Acute lymphoblastic leukemias in
3. Yang JJ, Cheng C, Yang W, et al: Genome-wide interrogation adults. In Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors:
of germline genetic variation associated with treatment Hematology: basic principles and practice, ed 5, Philadelphia,
response in childhood acute lymphoblastic leukemia. JAMA 2009, Churchill Livingstone, chapter 66.
301:393-403, 2009. 14. Attarbaschi A, Mann G, Dworzak M, et al: Mediastinal mass in
4. National Cancer Institute: Childhood acute lymphoblastic childhood T-cell acute lymphoblastic leukemia: significance
leukemia treatment (PDQ), updated June 20, 2010. Avail- and therapy response. Med Pediatr Oncol 39:558-565, 2002.
able at: http://www.cancer.gov/cancertopics/pdq/treatment/ 15. Goldberg JM, Silverman LB, Levy DE, et al: Childhood acute
childALL/HealthProfessional. Accessed July 6, 2010. lymphoblastic leukemia: the Dana Farber Cancer Institute
5. Acute lymphoblastic leukemia in children. National Cancer acute lymphoblastic leukemia consortium experience. J Clin
Institute Fact Sheet, reviewed July 11, 2002. Available at: Oncol 21:3616-3622, 2003.
http://www.cancer.gov/cancertopics/factsheet/sites-types/all- 16. The Leukemia and Lymphoma Society: Leukemia facts and
in-children. Accessed July 7, 2010. statistics. updated July 01, 2009. Available at: http://www.
6. Advani A, Jin T, Bolwell B, et al: A prognostic scoring system leukemia.org/all_page?item_id-9346. Accessed July 7, 2010.
for adult patients less than 60 years of age with acute lym- 17. Kalina T, Vaskova M, Mejstrikova E, et al: Myeloid antigens in
phoblastic leukemia in first relapse. Leuk Lymphoma 50:1126- childhood lymphoblastic leukemia: clinical data point to
1131, 2009. regulation of CD66c distinct from other myeloid antigens.
7. Pui C-H, Evans WE: Treatment of acute lymphoblastic leuke- BMC Cancer 5:38, 2005.
mia. N Engl J Med 354:166-178, 2006. 18. Pilozzi E, Pulford K, Jones M, et al: Co-expression of CD
8. Swerdlow SH, Campo E, Harris NL, et al, editors: WHO 79a(JCB117) and CD3 by lymphoblastic leukemia. J Pathol
classification of tumours of haematopoietic and lymphoid tissues, 186:140-143, 1998.
ed 4, Lyon, France, 2008, IARC Press. 19. Greaves MF, Wiemels J: Origins of chromosome transloca-
9. Hutter JJ: Childhood Leukemia. Pediatr Rev 31:234-241, 2010. tions in childhood leukaemia. Nat Rev Cancer 3:639-649.
Also available at: http://pedsinreview.aappublications. 2003.
org/cgi/content/full/31/6/234. Accessed_Sept. 9, 2010. 20. Castor A, Nilsson L, Astrand-Grundstrm I, et al: Distinct
10. Onciu M: Acute lymphoblastic leukemia. Hematol Oncol Clin patterns of hematopoietic stem cell involvement in acute
North Am 23:655-674, 2009. lymphoblastic leukemia. Nat Med 11:630-637, 2005.
CHAPTER 36 Acute Leukemias 557
21. Najfeld V: Conventional and molecular cytogenetic basis of 33. Nasr R, Lallemand-Breitenbach V, Zhu J, et al: Therapy-
hematologic malignancies. In Hoffman R, Benz EJ. Jr, Shattil induced PML/RARA proteolysis and acute promyelocytic leu-
SJ, et al, editors: Hematology: basic principles and practice, ed 5, kemia cure. Clin Cancer Res 15(20):6321-6326, 2009.
Philadelphia, 2009, Churchill Livingstone, chap 55. 34. Duchayne E, Fenneteau O, Pages M-P, et al: Acute megakaryo-
22. Brugnoletti F, Morris EB, Laningham FH, et al: Recurrent blastic leukemia: a national clinical and biological study
intrathecal methotrexate induced neurotoxicity in an adoles- of 53 adult and childhood cases by the Groupe Franais
cent with acute lymphoblastic leukemia: Serial clinical and dHmatologie Cellulaire (GFHC). Leuk Lymphoma 44:49-58,
radiologic findings. Pediatr Blood Cancer 52:293-295, 2009. 2003.
23. Angiolillo AL, Yu AL, Reaman G, et al: A phase II study of 35. Weinberg OK, Seetharam M, Ren L, et al: Clinical character-
Campath-1H in children with relapsed or refractory acute ization of acute myeloid leukemia with myelodysplasia-
lymphoblastic leukemia: a Childrens Oncology Group related changes as defined by the 2008 WHO classification
report. Pediatr Blood Cancer 53:978-983, 2009. system. Blood 113:1906-1908, 2009.
24. Uchiumi H, Matsushima T, Yamane A, et al: Prevalence and 36. Ngo N, Lampert IA, Naresh KN: Bone marrow trephine find-
clinical characteristics of acute myeloid leukemia associated ings in acute myeloid leukemia with multilineage dysplasia.
with disseminated intravascular coagulation. Int J Hematol Br J Haematol 140(3):279-286, 2008.
86(2):137-142, 2007. 37. Czader M, Orazi A: Therapy related myeloid neoplasms. Am
25. Miller KB, Pihan G: Clinical manifestations of acute myeloid J Clin Pathol 132:410-425, 2009.
leukemia. In Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: 38. Vardiman JW: The World health Organization (WHO) clas-
Hematology: basic principles and practice, ed 5, Philadelphia, sification of tumors of the hematopoietic and lymphoid
2009, Churchill Livingstone. tissues: an overview with emphasis on the myeloid neo-
26. Montesinos P, Lorenzo I, Martn G, et al: Tumor lysis syn- plasms. Chem Biol Interact 184(1-2):16-20 (review), 2010.
drome in patients with acute myeloid leukemia: identifica- Epub October 24, 2009.
tion of risk factors and development of a predictive model. 39. Anderson LA, Pfeiffer RM, Landgren O, et al: Risks of myeloid
Haematologica 93(1):67-74, 2008. malignancies in patients with autoimmune conditions. Br J
27. Cairo MS, Bishop M: Tumour lysis syndrome: new therapeu- Cancer 100:822-828, 2009.
tic strategies and classification. Br J Haematol 127:3-11, 2004. 40. Bennett JM, Catovsky D, Daniel MT, et al: Proposals for the
28. Larson RA, Hall MJ: Acute leukemia. In Hall JB, Schmidt classification of the acute leukemias. French-American-British
GA, Wood LDH, editors: Principles of critical care, ed 3, New (FAB) co-operative group. Br J Haematol 33:451-458, 1976.
York, 2005, McGraw-Hill Medical. Available at: http://www. 41. Bennett JM, Catovsky D, Daniel MT, et al: Proposed revised
accessmedicine.com/content.aspx?aID=2293953. Accessed criteria for the classification of acute myeloid leukemia. A
Sept. 6, 2010. report of the French-American-British Cooperative Group.
29. Kelly LM, Filliland DG: Genetics of myeloid leukemias. Annu Ann Intern Med 103:620-625, 1985.
Rev Genomic Hum Genet 3:179-198 (review), 2002. 42. Kaleem Z, Crawford E, Pathan H, et al: Flow cytometric analy-
30. Heerema-McKenney A, Arber DA: Acute myeloid leukemias. sis of acute leukemias. Arch Pathol Lab Med 127:42-48, 2003.
Hematol Oncol Clin North Am 23:633-654, 2009. 43. Villeneuve P, Kim DT, Xu W: The morphological subcatego-
31. Mrzek K, Marcucci G, Paschka P, et al: Advances in molecu- ries of acute monocytic leukemia (M5a and M5b) share
lar genetics and treatment of core-binding factor acute similar immunophenotypic and cytogenetic features and
myeloid leukemia. Curr Opin Oncol 20(6):711-718 (review), clinical outcomes. Leuk Res 32:269-273, 2008.
2008. 44. Xavier AC, Ge Y, Taub JW: Down syndrome and malignancies:
32. Stein E, McMahon B, Kwaan H, et al: The coagulopathy of a unique clinical relationship: a paper from the 2008 William
acute promyelocytic leukemia revisited. Best Pract Res Clin Beaumont Hospital Symposium on Molecular Pathology. J
Haematol 22(1):153-163 (review), 2009. Mol Diagn 11:371-380, 2009.
37 Mature Lymphoid Neoplasms
Magdalena Czader

OUTLINE OBJECTIVES
Morphologic and After completion of this chapter, the reader will be able to:
Immunophenotypic
Features of Normal 1. Describe normal lymph node morphology and discuss 5. Discuss the most commonly occurring mature B-
Lymph Nodes the function of various compartments and constituent and T-cell neoplasms, including epidemiology, clinical
Cortex cells. presentation, pathophysiology, lymph node histologic
Paracortex 2. Outline the most common histologic patterns of reac- features, peripheral blood or bone marrow findings,
Medulla tive lymphadenopathies. and diagnostic test results.
Sinuses 3. Describe the peripheral blood findings in chronic lym- 6. Interpret diagnostic test results to identify lymphopro-
Lymph Node Processing phocytic leukemia and hairy cell leukemia. liferative disorders.
Reactive 4. Describe the approach to the diagnosis of lympho-
Lymphadenopathies mas as outlined by the World Health Organization
Follicular Pattern
classification.
Paracortical Pattern
Sinusoidal Pattern
Mixed Pattern
Lymphomas
Mature B-Cell
Lymphomas
Mature T-Cell and Natural
Killer Cell Lymphomas
Hodgkin Lymphoma

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 46-year-old, previously healthy man came for evaluation B), CD10, and BCL-6 positivity, and focal CD30 antigen
of an enlarged left cervical lymph node. The patient had expression.
discovered this isolated lymphadenopathy 2 weeks previ- 1. What is your diagnosis based on the histologic and immu-
ously and did not complain of any other symptoms. The nophenotypic features?
lymph node measured approximately 2cm. The findings of 2. What additional immunophenotypic features that confirm
his physical examination were otherwise unremarkable. The the diagnosis could be seen using flow cytometry?
lymph node was excised, and microscopic examination 3. Is it likely that this patient would show disseminated
showed the histologic features presented in Figure 37-1, A. disease, including bone marrow involvement?
Immunohistochemical stains showed CD20 (see Figure 37-1, Continued

558
CHAPTER 37 Mature Lymphoid Neoplasms 559

CASE STUDYcontd
After studying the material in this chapter, the reader should be able to respond to the following case study:

A B
Figure 37-1 Histologic lymph node findings for the patient in the case study. A, Lymph node (hematoxylin and eosin stain, 500). B, CD20 antigen
expression in the lymphoid population (immunoperoxidase stain, 500).

L
ymphomas are neoplastic lesions of the lymphoid
system. Original microscopic observations and, more Subcapsular sinus
recently, immunophenotypic and molecular studies
Primary and secondary
have confirmed that these neoplasms recapitulate specific follicles
stages of differentiation of normal lymphoid cells. The diagno-
Cortex
sis is based on the combination of biologic features such as
Paracortex
morphology, immunophenotype and molecular genetic char-
Medulla
acteristics, and clinical information.1 Thus, during initial
sample processing the appropriate steps must be taken to
ensure tissue preservation and availability for microscopic
Efferent lymphatic
examination and immunophenotypic and molecular studies.
Knowledge of normal lymphoid differentiation is a pre
requisite for understanding the lymphoid neoplasms. This
Capsule
chapter describes the morphologic and immunophenotypic
features of normal lymph nodes and selected common lym-
phomas and lymphoproliferative disorders. Reactive lymphoid Afferent lymphatics
hyperplasias, which can resemble neoplastic lesions, also are
discussed.
Figure 37-2 Diagram of a normal lymph node showing cortical, paracorti-
cal, and medullary compartments.
MORPHOLOGIC AND IMMUNOPHENOTYPIC
FEATURES OF NORMAL LYMPH NODES
mucosa-associated lymphoid tissue (MALT), which is the primary
Lymphoid organs serve as sites of antigen recognition, antigen site of antigenic contact at these locations; these aggregates
processing, and lymphopoiesis. Most of the lymphoid tissue is drain directly into regional lymph nodes.
concentrated in lymph nodes, which are round to oval encap- Histologic components of the lymph node include the
sulated organs serving as primary sites of immunologic cortex, paracortex, medullary cords, and sinuses (Figure 37-2).
response. They are particularly prominent at sites with an envi- These are not only structural but also functional compartments
ronmental interface. Large groups of lymph nodes are found serving as sites of immunologic reactions for specific antigenic
draining specific peripheral areas (e.g., cervical, axillary, or stimuli.
inguinal). Similarly, internal organs are served by regional
lymph nodes (e.g., mediastinal, hilar, and mesenteric). Respi- Cortex
ratory and digestive tracts have additional aggregates of The lymph node is surrounded by a capsule of fibrous tissue.
lymphoid tissue located directly in the mucosa called Immediately below the capsule is the cortex, the most
560 PART V Leukocyte Disorders

Plasma cell
CD19-
B-immunoblast
CD20-
CD19+
CD38+
CD20+
CD138+
slg+
Naive B-cell slg+
CD19+
CD20+
CD5+
B-cell of Centroblast Centrocyte
primary follicle
CD19+ CD19+ Germinal centers CD19+
CD20+ CD20+
CD20+ CD10+
CD5 +/- CD10+
BCL-6+ BCL-6+
BCL-2- BCL-2-
slg- slg+

Memory B cell
CD19+
CD20+
BCL-6-
BCL-2+
slg+

Figure 37-3 Differentiation stages of mature B cells. Note changes in


immunophenotype at specific stages of differentiation. BCL, B-cell lym-
phoma; sIg, surface immunoglobin.

superficial portion of the lymph node consisting of primary


and secondary follicles. Primary follicles are microscopic aggre-
gates of small, round naive B lymphocytes. These lymphocytes
express panB-cell markers, including CD19 and CD20, and
are frequently CD5+ (Figure 37-3). The formation of secondary
follicles, including germinal centers, starts with antigen presen-
Figure 37-4 Secondary follicle with well-developed polarization in germi-
tation by follicular dendritic cells.2,3 On antigen encounter, nal center showing dark zone (DZ) and light zone (LZ). Note the presence of
naive B lymphocytes undergo transformation, proliferation, numerous tingible body macrophages (arrows). A distinct mantle zone (MZ)
and differentiation into precursors of antibody-producing also is present at the periphery of the germinal center (hematoxylin and eosin
plasma cells and memory B cells (see Figure 37-3). The remain- stain, 100).
ing naive B cells are displaced into the periphery of the germi-
nal center and form the mantle zone.
Germinal center B cells have a specific immunophenotype. into memory B cells that form a marginal zone at the
In addition to panB-cell markers, they express germinal center periphery of the mantle zone. Marginal zone lymphocytes are
cell antigens CD10 and BCL-6, and, in contrast to circulating medium-sized with abundant clear cytoplasm and indented
B cells, they lack antiapoptotic BCL-2 protein. The functional nuclei.
compartments of the germinal center include the dark zone The final step of B-cell differentiation is plasma cells that
occupied by centroblasts, large B cells with round vesicular are present in the medullary cords of the lymph nodes and
nuclei, small nucleoli adjacent to nuclear membrane, and migrate to the bone marrow. Plasma cells are negative for pan
basophilic cytoplasm (Figure 37-4). The dark zone is a site of B-cell antigens and surface immunoglobulins; however, they
high proliferative activity and somatic mutations of B-cell express CD138, CD38, and cytoplasmic immunoglobulins.
immunoglobulin variable regions. The latter process allows for
the production of immunoglobulins with the best affinity for Paracortex
a particular antigen. The paracortex occupies the area separating the follicles and
After completing somatic mutations, centroblasts differen- extends toward the medullary cords. This compartment gener-
tiate into centrocytes, smaller cells with dense chromatin ates immunocompetent T cells and is occupied predominantly
and irregular nuclear outlines, which form the light zone by T cells, interdigitating dendritic cells (antigen-presenting
(see Figure 37-4). Subsequently, centrocytes with low-affinity cells), and high endothelial venules. The latter are specialized
(unfit) surface immunoglobulins undergo apoptosis and are vessels serving as a gate of entry for lymphocytes from periph-
phagocytized by germinal center macrophages (tingible body eral blood into the lymph node. T cells express panT-cell
macrophages). The presence of numerous macrophages with antigens CD3, CD5, CD2, and CD7. Both CD4+ and CD8+ T
apoptotic debris contributes to the characteristic starry sky lymphocytes are seen in the paracortex. Similar to B cells, T
pattern of the germinal center. Centrocytes with immuno cells transform in response to antigen stimulation. In this
globulins with high affinity for a particular antigen lose their process, small lymphocytes become immunoblasts: large lym-
germinal center antigens (CD10 and BCL-6) and differentiate phoid cells with vesicular nuclei; prominent nucleoli, often
CHAPTER 37 Mature Lymphoid Neoplasms 561

single; and abundant basophilic cytoplasm. The paracortex


REACTIVE LYMPHADENOPATHIES
also contains numerous B immunoblasts.
Lymphadenopathy can occur in benign and malignant condi-
Medulla tions. Any antigenic stimulation can result in lymph node
The medulla represents the innermost portion of the lymph enlargement. Reactive lymphadenopathies can affect any com-
node surrounding the hilum. This area is composed of medul- partment of the lymph node and present as selective expansion
lary cords with plasma cells and medullary sinuses. of normal nodal structures. Reactive hyperplasias are classified
into several patterns, as follows:
Sinuses 1. Follicular
The filtration of lymphatic fluid through the lymph node is 2. Paracortical
accomplished through afferent lymphatics communicating 3. Sinusoidal
with the subcapsular sinus, which is situated immediately 4. Mixed
beneath the capsule (see Figure 37-2). The subcapsular sinus
drains to cortical sinuses, which run through the cortex and Follicular Pattern
empty to medullary sinuses. The latter converge into the effer- Follicular hyperplasia is the most common of the reactive
ent lymphatic vessel at the hilum. The sinuses are filled with lymphadenopathies. It is seen frequently in lymph nodes and
macrophages or sinus histiocytes. These cells play an important tonsils of children and adolescents as a reaction to infections.
role in antigen capture and processing. In adults, it occurs in association with autoimmune disorders
(rheumatoid arthritis, systemic lupus erythematosus), syphilis,
and early human immunodeficiency virus (HIV) infection.
LYMPH NODE PROCESSING
Microscopically, the expansion of reactive follicles can be
The current approach to the diagnosis of lymphomas incorpo- prominent and extend beyond the cortex into the medulla
rates routine light microscopic examination and ancillary tech- (Figure 37-5). The follicles retain all the hallmarks of reactive
niques. During sample processing, appropriate steps should be germinal centers, including distinct polarization, presence of
taken to ensure adequate preservation of the tissue and its tingible body macrophages, abundant mitotic figures, and a
availability for all necessary studies. The appropriate transport preserved mantle zone (see Figure 37-4).
conditions need to be maintained to preserve tissue integrity
and prevent drying. Immediately after excision, the lymph Paracortical Pattern
node should be transported to the pathology laboratory in a Paracortical expansion is associated with viral infections (e.g.,
sealed jar on gauze pads moistened with sterile saline. The infectious mononucleosis) and drug reactions and is also seen
fresh lymph node is cut into 3-mm-thick sections for the evalu- in patients with chronic skin diseases (dermatopathic lymph-
ation of nodal architecture. If areas of granulomas or suppura- adenopathy). In addition to small lymphocytes, the paracortex
tion are present, a portion of the tissue should be sent for shows numerous immunoblasts, increased mitotic activity,
cultures. and vascular proliferation (Figure 37-6). Focal areas of necrosis
Touch imprints can be prepared to ensure the adequacy may also be seen. In dermatopathic lymphadenopathy, the
of the specimen and to perform special studies. To obtain paracortex has a characteristic mottled appearance as a result
an adequate imprint, a freshly cut tissue surface is gently
touched to the glass slide and pulled away. Touch imprints
can be fixed in formalin or alcohol solution or air-dried for
subsequent Wright-Giemsa staining. Storing of fixed touch
imprints for immunocytochemical studies is optional, because
currently immunophenotyping is most commonly performed
on paraffin-embedded tissue or using flow cytometry. The
latter is particularly helpful in confirming monoclonal light
chain expression.
Several thin sections of a lymph node are placed in 10%
buffered formalin for paraffin embedding. Some pathology
laboratories fix additional tissue samples in a variety of fixa-
tives with protein-precipitating properties (B5 fixative, zinc
chloride formalin) for better preservation of cytologic detail.4
Regardless of the fixative used, thin sectioning of the fresh
lymph node is crucial for the proper permeation of the tissue.
A portion of the lymph node is placed in culture medium
(Roswell Park Memorial Institute medium) and transported to
the flow cytometry laboratory for immunophenotyping. The Figure 37-5 Reactive follicular hyperplasia with numerous secondary
remaining fresh tissue can be stored at 70 C for further follicles scattered throughout the lymph node (hematoxylin and eosin stain,
studies. 40).
562 PART V Leukocyte Disorders

Figure 37-6 Immunoblasts (arrows) scattered in the paracortex (hema- Figure 37-8 Dermatopathic lymphadenopathy in a patient with chronic
toxylin and eosin stain, 1000). skin rash. Scattered pigment-laden macrophages (arrow) are present in the
paracortical area (hematoxylin and eosin stain, 400).

A
Figure 37-7 Paracortical hyperplasia. Note mottled appearance of the
paracortex resulting from multiple scattered histiocytes with abundant cyto-
plasm (arrows) (hematoxylin and eosin stain, 40).

of an increased number of large cells with abundant clear


cytoplasm scattered among small lymphoid cells (Figure 37-7).
These cells include histiocytes, often carrying melanin pigment,
and Langerhans cells (Figure 37-8). Scattered immunoblasts,
plasma cells, eosinophils, and vascular proliferation are also
encountered.

Sinusoidal Pattern
Expanded subcapsular, cortical, and medullary sinuses are
often seen in lymph nodes draining limbs, abdominal organs,
various inflammatory lesions, and malignancies. In advanced B
cases, the prominent sinuses compress the nodal parenchyma.
Figure 37-9 Toxoplasma lymphadenitis. A, Monocytoid B cells are
They may be completely filled with histiocytes showing abun-
medium-sized with irregular nuclear outlines and abundant cytoplasm (shown
dant cytoplasm, a small oval nucleus with inconspicuous in center circle) (hematoxylin and eosin stain, 400). B, Classic triad of
nucleolus, and delicate chromatin. Monocytoid B cells with reactive changes seen in Toxoplasma lymphadenitis includes follicular hyper-
abundant cytoplasm and oval indented nuclei that may mimic plasia with focal aggregates of histiocytes (H) and monocytoid B cells (MBC).
histiocytes are seen in nodal sinuses in HIV-associated lymph- An irregular outline of a secondary follicle is seen on the left (arrowheads)
adenopathy and Toxoplasma lymphadenitis (Figure 37-9, A). (hematoxylin and eosin stain, 100).
CHAPTER 37 Mature Lymphoid Neoplasms 563

Numerous malignant lesions show a predilection for sinuses, immunoglobulins, clonal immunoglobulin gene rearrange-
such as Langerhans cell histiocytosis, B-cell and T-cell lympho- ments, or both. Follicular lymphoma and diffuse large B-cell
mas, and carcinomas; a high-power microscopic evaluation of lymphoma (DLBCL) are the most common subtypes of B-cell
expanded sinuses is always necessary. lymphoma.9 Most cases are lymph node based and occur in
elderly individuals. However, leukemic involvement (periph-
Mixed Pattern eral blood and bone marrow) can occur with any lymphoma
A classic example of mixed-pattern hyperplasia is seen in Toxo- subtype. The most common mature B-cell neoplasms are dis-
plasma gondii infection, a common protozoal infection typi- cussed in the following paragraphs and are summarized in
cally seen after ingestion of raw meat or contamination by cat Table 37-2.
feces. Histologically, the expansion of all lymph node compart-
ments is seen (see Figure 37-9). Florid follicular hyperplasia is Chronic Lymphocytic Leukemia/Small
accompanied by paracortical expansion, aggregates of histio- Lymphocytic Lymphoma
cytes encroaching on germinal centers, and expanded sinuses. Definition. Chronic lymphocytic leukemia (CLL) and
Sinuses are focally filled with a specific subset of B cells, small lymphocytic lymphoma (SLL) are characterized by the
so-called monocytoid B cells. accumulation of small lymphoid cells in the peripheral blood,
bone marrow, and lymphoid organs. They are derived from
recirculating B cells expressing CD5, immunoglobulin M
LYMPHOMAS
(IgM), and IgD that are normally present in the peripheral
Approximately 86,000 new cases of lymphoma are diagnosed blood. The World Health Organization (WHO) classification
annually in the United States.5 Most lymphomas develop considers CLL and SLL as one entity with different clinical
in previously healthy individuals. The strongest risk factor presentations. The diagnosis is based on the predominant site
for development of lymphoproliferative disorder is altered of involvement. CLL, by definition, has involvement of the
immune function as seen in immunocompromised patients or peripheral blood (monoclonal B-lymphocytes greater than
individuals with autoimmune disease.6,7 Similarly, certain viral 5 109 cells/L) and bone marrow. SLL primarily involves
and bacterial infections are associated with a higher risk for the lymph nodes and other lymphoid organs.
development of lymphoma.8 Accumulating evidence indicates
that exposure to chemicals and herbicides may predispose to Morphology and Immunophenotype. Bone marrow
lymphoid neoplasms. Most lymphomas present in lymph and peripheral blood films shows small lymphoid cells with a
nodes. Certain types show a predilection for extranodal sites. coarse chromatin pattern, inconspicuous nucleoli, and scant
The frequency of bone marrow and peripheral blood (leukemic cytoplasm10 (Figure 37-10). Larger lymphoid cells with less
phase) involvement varies depending on the lymphoma condensed chromatin and distinct nucleoli (prolymphocytes)
subtype. are rare. Smudge cells, representing disintegrated lymphoid
Over the years, numerous classifications have been pro- cells, are present on the peripheral blood film. These cells are
posed based mainly on the morphology and clinical character- helpful in the diagnosis because they are not often seen in
istics (e.g., Rappaport classification, Kiel system, Working other subtypes of malignant lymphoma. The bone marrow
Formulation). With increased understanding of the develop- biopsy specimen shows nodular, diffuse, or interstitial infil-
ment and function of the immune system, however, it became trates of small lymphoid cells (Figure 37-11).
clear that lymphomas, like myeloid neoplasms, recapitulate Lymph nodes involved by SLL show an effacement of
normal stages of lymphoid differentiation. In addition, the normal nodal architecture (Figure 37-12, A) by a diffuse
elucidation of specific molecular events occurring in lymphom-
agenesis helped in devising a clinically relevant subclassifica-
tion, especially for morphologically heterogeneous entities.
Currently, numerous subtypes of lymphoma are distinguished
based on morphology, immunophenotype, molecular genetics,
and clinical characteristics. The integration of these features is
mandatory for comprehensive lymphoma diagnosis. On the
basis of cellular origin, lymphomas can be categorized into
lesions of lymphoid precursors and neoplasms of mature lym-
phoid cells (Table 37-1). In this chapter, only mature B-cell
and T-cell neoplasms are discussed; the precursor malignancies
are covered in Chapter 36.

Mature B-Cell Lymphomas


Mature B-cell lymphomas are neoplasms derived from
various stages of B-cell differentiation. Although they show Figure 37-10 Chronic lymphocytic leukemia/small lymphocytic lym-
significant morphologic and immunophenotypic heterogene- phoma. Peripheral blood film showing small lymphocytes and smudge cells
ity, all B-cell lymphomas produce monoclonal light chain (arrows) (Wright-Giemsa stain, 1000).
564 PART V Leukocyte Disorders

TABLE 37-1 2008 World Health Organization Classification of Mature Lymphoid Neoplasms
Type of Lymphoma Examples
Mature B-cell lymphomas Chronic lymphocytic leukemia/small lymphocytic lymphoma
B-cell prolymphocytic leukemia
Splenic B-cell marginal zone lymphoma
Hairy cell leukemia
Splenic B-cell lymphoma/leukemia, unclassifiable
Splenic diffuse red pulp small B-cell lymphoma
Hairy cell leukemia-variant
Lymphoplasmacytic lymphoma
Heavy chain diseases
Gamma Heavy chain disease
Mu Heavy chain disease
Alpha Heavy chain disease
Plasma cell neoplasms
Monoclonal gammopathy of undetermined significance (MGUS)
Plasma cell myeloma
Solitary plasmacytoma of bone
Extraosseous plasmacytoma
Monoclonal immunoglobulin deposition diseases
Extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma)
Nodal marginal zone lymphoma
Follicular lymphoma
Primary cutaneous follicle center lymphoma
Mantle cell lymphoma
Diffuse large B-cell lymphoma (DLBCL), not otherwise specified
T-cell/histiocyte-rich large B-cell lymphoma
Primary DLBCL of the central nervous system
Primary cutaneous DLBCL, leg type
Epstein-Barr virus (EBV)positive DLBCL of the elderly
DLBCL associated with chronic inflammation
Lymphomatoid granulomatosis
Primary mediastinal (thymic) large B-cell lymphoma
Intravascular large B-cell lymphoma
ALK-positive large B-cell lymphoma
Plasmablastic lymphoma
Large B-cell lymphoma arising in human herpesvirus 8associated multicentric Castleman disease
Primary effusion lymphoma
Burkitt lymphoma
B-cell lymphoma, unclassifiable, with features intermediate between those of DLBCL and Burkitt lymphoma
B-cell lymphoma, unclassifiable, with features intermediate between those of DLBCL and classical Hodgkin lymphoma
Mature T-cell lymphomas T-cell prolymphocytic leukemia
T-cell large granular lymphocytic leukemia
Chronic lymphoproliferative disorder of natural killer (NK) cells
Aggressive NK cell leukemia
EBV-positive T-cell lymphoproliferative diseases of childhood
Systemic EBV-positive T-cell lymphoproliferative disease of childhood
Hydroa vacciniformelike lymphoma
Adult T-cell leukemia/lymphoma
Extranodal NK/T-cell lymphoma, nasal type
Enteropathy-associated T-cell lymphoma
Hepatosplenic T-cell lymphoma
Subcutaneous panniculitis-like T-cell lymphoma
Mycosis fungoides
Szary syndrome
Primary cutaneous CD30+ T-cell lymphoproliferative disorders
CHAPTER 37 Mature Lymphoid Neoplasms 565

TABLE 37-1 2008 World Health Organization Classification of Mature Lymphoid Neoplasmscontd
Type of Lymphoma Examples
Primary cutaneous peripheral T-cell lymphomas, rare subtypes
Primary cutaneous gamma-delta T-cell lymphoma
Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma
Primary cutaneous CD4+ small/medium T-cell lymphoma
Peripheral T-cell lymphoma, not otherwise specified
Angioimmunoblastic T-cell lymphoma
Anaplastic large cell lymphoma, ALK positive
Anaplastic large cell lymphoma, ALK negative

TABLE 37-2 Morphologic and Immunophenotypic Features of Mature B-Cell Lymphomas


Immunophenotype/
Subtype Architectural Features Cytologic Characteristics Cytogenetics Cell of Origin
Chronic lymphocytic Diffuse lymphocytic Small lymphoid cells CD20+, CD19+, CD5+, CD23+ Naive or memory
leukemia/small lymphocytic proliferation with growth B cells
lymphoma centers
B-cell prolymphocytic Diffuse proliferation Medium-sized lymphoid cells with CD20+, CD19+, FMC7+, CD5+/ Unknown mature
leukemia distinct punched-out nucleoli B cell
and abundant cytoplasm
Mantle cell lymphoma Diffuse, nodular, or mantle Medium-sized lymphocytes with CD20+, CD19+, CD5+, FMC7+, Mantle zone cell
zone pattern irregular nuclei cyclin D1+, t(11;14)
Follicular lymphoma Follicular pattern Medium-sized lymphocytes with CD20+, CD19+, CD10+, Germinal center
indented nuclei and variable BCL-6+, BCL-2+ cell
numbers of large lymphoid t(14;18)
cells
Extranodal marginal zone Diffuse lymphoid proliferation, Medium-sized lymphocytes with CD20+, CD19+, CD43+/ Marginal zone cell
lymphoma of mucosa- occasionally marginal zone irregular nuclei and clear
associated lymphoid tissue or nodular pattern abundant cytoplasm
Plasma cell myeloma, Sheets or large aggregates of Plasma cells, frequently with CD20, CD19+/, CD38+, Plasma cell
plasmacytoma plasma cells cytologic atypia CD138+, cytoplasmic
light chain+
Diffuse large B-cell lymphoma Diffuse proliferation Large lymphoid cells CD20+, CD19+, CD10+/, Different stages of
BCL-6+/, BCL-2+/, CD5+/ mature B cells
Burkitt lymphoma Diffuse lymphoid proliferation Medium-sized lymphocytes with CD20+, CD19+, CD10+, Germinal center
with starry sky pattern evenly distributed chromatin, BCL-6+, BCL-2, t(8:14) cell
inconspicuous nucleoli

proliferation of small round lymphoid cells with coarse chro- postgerminal center stage), with mutated VH genes. Both types
matin, indistinct nucleoli, and scant cytoplasm. In addition, are positive for panB-cell antigens, including CD19 and
scattered nodules (so-called pseudofollicles, growth centers, or weakly expressed CD20. In addition, the expression of CD5,
proliferation centers) composed of medium-sized and large CD23, and weak surface monoclonal or light chains is seen.
lymphoid cells with dispersed chromatin and distinct nucleoli The presence of CD23 and absence of FMC7 and cyclin D1
are observed (see Figure 37-12, B). The diffuse proliferation of distinguish CLL/SLL from mantle cell lymphoma. The presence
small lymphoid cells with pseudofollicles is pathognomonic of monoclonal B cells with an immunophenotype similar to
for SLL. that of CLL/SLL has been described in a small proportion of
As noted earlier, CLL/SLL is derived from circulating healthy individuals; thus the demonstration of these cells by
CD5+IgM+IgD+ B cells. Currently, two groups of CLL/SLL are flow cytometry must always to be interpreted in the context of
recognized.11 The first corresponds to the pregerminal center other clinical and laboratory features.12
phenotype with naive B cells showing no mutations in the
variable region of the immunoglobulin heavy chain (VH) gene. Clinical Features and Prognosis. Overall, CLL/SLL
The second form is derived from memory B cells (the is regarded as an indolent lymphoproliferative disorder of
566 PART V Leukocyte Disorders

A
Figure 37-11 Chronic lymphocytic leukemia/small lymphocytic lym-
phoma. Bone marrow biopsy specimen with nodular (right) and interstitial
lymphoid infiltrate composed of small lymphocytes (hematoxylin and eosin
stain, 400).

the elderly (median age at diagnosis is 65 years). Although


this disease is incurable by current therapeutic approaches,
the median survival is 10 years.13 A variety of features are
used to predict patient outcome. Commonly used clinical
staging systems are based on the extent of lymphoid organ
involvement and degree of cytopenias. In addition, distinct
cytogenetic abnormalities that roughly correspond to naive
and memory phenotypes (unmutated and mutated VH gene)
are of prognostic significance.11 Trisomy of chromosome 12
is seen predominantly in cases of pregerminal center origin B
and correlates with a rapidly progressive clinical course. Figure 37-12 Chronic lymphocytic leukemia/small lymphocytic lym-
These patients may require aggressive treatment early in the phoma in a lymph node. A, Diffuse proliferation of small lymphoid cells
course of the disease. In contrast, the deletion of 13q14 (hematoxylin and eosin stain, 100). B, Proliferation center with large and
is associated with postgerminal center (memory) phenotype medium-sized lymphoid cells in a background of small lymphocytes (hema-
and predicts a more indolent clinical course. Independent toxylin and eosin stain, 400).
of the course of the original disease, approximately 5% of
patients with CLL/SLL develop a DLBCL (so-called Richter
transformation).

Prolymphocytic Leukemia
Definition. Prolymphocytic leukemia (PLL) is a rare
mature lymphoid leukemia that can be derived from B or T
cells. Both B-cell and T-cell types involve peripheral blood,
bone marrow, and spleen. Lymph node involvement is more
commonly seen in T-cell PLL. This lymphoproliferative disease
is distinct from CLL, and its diagnosis requires that more than
55% of circulating lymphoid cells have the morphology of a
prolymphocyte.

Morphology and Immunophenotype. The pathogno-


monic cell of B-cell PLL is a prolymphocyte of medium size
with round nucleus, moderately abundant cytoplasm, and dis-
tinct punched-out nucleolus (Figure 37-13). The cell size
(twice that of a normal lymphocyte) and prominent central Figure 37-13 Peripheral blood film showing a characteristic morphology
nucleolus allow PLL to be distinguished from CLL/SLL. The of prolymphocytes (medium size, abundant cytoplasm, distinct nucleoli)
peripheral blood involvement in PLL is typically prominent (1000).
CHAPTER 37 Mature Lymphoid Neoplasms 567
with a white blood cell count in excess of 100 109/L. Bone
marrow shows interstitial and/or nodular proliferation of pro-
lymphocytes. Both white and red splenic pulp are infiltrated
by PLL. B-cell PLL is positive for panB-cell markers CD20,
CD19, CD22, and FMC7. The density of CD20 and surface
light chain is higher than in typical cases of CLL/SLL. A propor-
tion of cases are positive for CD5 antigen. In such cases, dis-
tinguishing the PLL from mantle cell lymphoma presenting
with a leukemic involvement might be challenging and requires
cytogenetic or molecular studies to exclude the presence of
t(11;14) (refer to the section on mantle cell lymphoma).
The morphologic features of T-cell PLL are not as distinct as
those of the B-cell type. The neoplastic cells seen in peripheral
blood films are small to medium-sized with round or irregular
nuclei, the latter resembling Szary cells. In only a proportion A
of cases do the cells show prominent nucleoli. Cytoplasmic
blebbing is common. Bone marrow, spleen, lymph node, and
occasionally skin involvement is diffuse with accentuation in
nodal paracortical areas and around the vessels in dermis. The
diagnosis of T-cell PLL is challenging when attempted using
morphologic features alone. A combination of morphologic,
clinical, and immunophenotypic features is most helpful in
making the diagnosis. T prolymphocytes are positive for T-cell
markers such as CD3, CD2, and CD5. In contrast to other T-cell
lymphomas, T-cell PLL is positive for CD7 antigen. Most com-
monly CD4 antigen is expressed. A minority of cases can be
double positive for CD4 and CD8, or positive for CD8.

Clinical Features and Prognosis. Like CLL/SLL, PLL is


a disease of the elderly (mean age of presentation is 70 years). B
Overall prognosis is poor (median survival is 3 years for B-cell Figure 37-14 Hairy cell leukemia. A, Interstitial bone marrow infiltrate
PLL), partly due to the high incidence of mutation in the tumor composed of widely spaced lymphoid cells with abundant cytoplasm and
suppressor gene TP53 and the unavailability of targeted irregular nuclei (hematoxylin and eosin stain, 1000). B, Lymphoid cells of
therapy.14 T-cell PLL is an aggressive disease with a median hairy cell leukemia show characteristic cytoplasmic projections, which are
survival of 1 year when conventional chemotherapies are used. supported by the peripheral cytoplasmic network of polymerized actin
Recently, addition of monoclonal antibody therapy against (peripheral blood, Wright-Giemsa stain, 1000).
CD52 (alemtuzumab) has significantly improved treatment
response and survival of patients with T-cell PLL.
peripheral blood films. Neoplastic cells display an oval or
Hairy Cell Leukemia indented nucleus, abundant cytoplasm, and fine, hairlike cyto-
Definition. Hairy cell leukemia is characterized by small plasmic projections (see Figure 37-14, B).
B lymphocytes with abundant cytoplasm and fine (hairy) Typical cases of hairy cell leukemia show strong positivity
cytoplasmic projections. The postulated cell of origin is the for B-cell markers (CD19, CD20, CD22) coupled with bright
peripheral B cell of postgerminal center stage (memory B cell). expression of CD11c, CD25, CD103, tartrate-resistant acid
phosphatase (TRAP, demonstrated by immunohistochemical
Morphology and Immunophenotype. Hairy cell leuke- analysis or cytochemical stain), DBA-44, and annexin A1.
mia cells are found predominantly in bone marrow and the Annexin A1 is the most specific marker for hairy cell leukemia
red pulp of the spleen. A low number of neoplastic B cells are and can help in differentiating hairy cell leukemia from splenic
seen in peripheral blood. Lymph node involvement is rare. The marginal zone lymphoma. Immunohistochemical stains for
bone marrow infiltrates are interstitial and are composed of annexin A1, TRAP, and DBA44 can be used when only pre-
small to medium-sized lymphoid cells with abundant cyto- served tissues are available (formalin fixed bone marrow biopsy
plasm (Figure 37-14, A). The bone marrow involvement may or clot section, or other involved tissues).
be subtle with preservation of normal hematopoiesis. As a
result of the production of fibrogenic cytokines by the leukemic Clinical Features and Prognosis. Hairy cell leukemia
cells, a bone marrow biopsy specimen shows an increase in is a rare lymphoproliferative disorder occurring in middle-aged
reticulin fibers. The characteristic cytologic features of neoplas- individuals (median age, 55 years). The presenting signs
tic cells are best appreciated in bone marrow aspirate and include splenomegaly and pancytopenia. Durable remissions
568 PART V Leukocyte Disorders

can be achieved using interferon- or purine analogues. Con-


ventional lymphoma therapy is not effective.

Mantle Cell Lymphoma


Definition. Mantle cell lymphoma is a lymphoprolifera-
tive disorder characterized by medium-sized lymphoid cells
with irregular nuclear outlines derived from the follicular
mantle zone.15

Morphology and Immunophenotype. The main sites


of presentation are lymph nodes. Bone marrow, peripheral
blood, spleen, and gastrointestinal tract also are frequently
involved. Most commonly, lymph nodes show a replacement
of normal nodal architecture with a diffuse proliferation of
monotonous, medium-sized lymphoid cells with irregular A
nuclear outlines (Figure 37-15, A). Occasionally, lymph nodes
can demonstrate a vaguely nodular pattern or partial preserva-
tion of nodal architecture with a prominent thickening
of mantle zones (see Figure 37-15, B). Peripheral blood
involvement by mantle cell lymphoma may mimic PLL (see
Figure 37-15, C).
Like other B-cell lymphoproliferative disorders, mantle cell
lymphoma shows expression of panB-cell markers (CD19,
CD20) and high-density clonal surface light chains. There is
coexpression of CD5 antigen; however, CD23 antigen is absent.
In contrast to CLL/SLL, the expression of CD20 and light
chains is strong, and there is immunoreactivity for cyclin D1.
Cyclin D1 (BCL1) is a proto-oncogene involved in the regula-
tion of G1 to S phase progression. In mantle cell lymphoma,
this gene is constitutively expressed through its translocation
to the immunoglobulin heavy chain gene, t(11;14). This B
cytogenetic abnormality is a defining feature of mantle cell
lymphoma.

Clinical Features and Prognosis. Mantle cell lym-


phoma is an aggressive lymphoproliferative disorder, and
median survival time is 3 to 5 years. Most patients present with
disseminated disease, including bone marrow involvement.
This lymphoma is incurable with current protocols.

Follicular Lymphoma
Definition. Follicular lymphoma originates from ger
minal centers and in most cases recapitulates follicular
architecture.

Morphology and Immunophenotype. Numerous C


closely spaced follicles replace the normal nodal architecture
(Figure 37-16). The neoplastic proliferation may extend into Figure 37-15 Mantle cell lymphoma. A, Diffuse proliferation of medium-
the perinodal adipose tissue. In contrast to the reactive second- sized lymphoid cells with irregular nuclei (hematoxylin and eosin stain,
ary follicles, in follicular lymphoma the mantle zone and 1000). B, Mantle zone pattern. Regressed germinal center is seen in the
center (hematoxylin and eosin stain, 100), C, Circulating mantle cell lym-
polarization are not present. The neoplastic follicles are
phoma cells with a few showing indented nuclei reminiscent of the cleaved
composed of medium-sized lymphoid cells with angular or
nuclei seen in histologic sections (peripheral blood film, Wright-Giemsa stain,
indented nuclei cytologically similar to centrocytes, with a vari- 1000).
able admixture of large lymphoid cells. The latter resemble
centroblasts and show oval nuclei with vesicular chromatin
and multiple marginally located nucleoli. The relative propor-
tion of medium-sized and large lymphoid cells is of prognostic
CHAPTER 37 Mature Lymphoid Neoplasms 569

Figure 37-16 Follicular lymphoma. Nodular proliferation replacing normal


elements of lymph node architecture (hematoxylin and eosin stain, 40).

significance. Cases with high numbers of large cells show a


significantly more aggressive clinical course, similar to that of
DLBCL. Follicular lymphomas are graded by counting the
average number of large cells per high-power field.1 Two grades
are recognized, with grade 1-2 lymphomas showing rare scat-
tered large lymphocytes, and grade 3 follicular lymphomas
composed of numerous centroblasts.
The immunophenotype reflects the follicle center cell origin
of this disease. PanB-cell markers (CD19, CD20) are present
along with the coexpression of CD10, BCL-6, and clonal
surface immunoglobulin. In contrast to reactive follicles, neo- B
plastic cells express BCL-2 protein. This protein is responsible Figure 37-17 Mucosa-associated lymphoid tissue lymphoma. A, Hetero-
for the decreased sensitivity to apoptosis and allows the accu- geneous population of medium-sized lymphoid cells with abundant clear
mulation of neoplastic lymphocytes. The expression of BCL-2 cytoplasm and occasional large cells (hematoxylin and eosin stain, 500).
by follicular lymphoma cells is due to the t(14;18)(q32;q21), B, Lymphoepithelial lesion. Malignant lymphocytes invading glandular epi-
which places the BCL2 gene under a promoter of the immu- thelium (hematoxylin and eosin stain, 1000).
noglobulin heavy chain gene. This cytogenetic abnormality is
present in 95% of cases.16
syndrome, Hashimoto thyroiditis) or previous infections (Heli-
Clinical Features and Prognosis. The median age at cobacter pylori gastritis or hepatitis C).17 Persistent immune
diagnosis is 59 years. Most patients present with disseminated stimulation leads to the accumulation of reactive lymphoid
disease. Bone marrow involvement is present in approximately tissue and subsequently to the development of marginal zone
50% of cases. The course of the disease is indolent in grade 1-2 lymphoma. The importance of continuous antigenic stimula-
follicular lymphoma, whereas grade 3 cases are more aggressive tion in the early stages of these lymphomas is shown by the
and are treated in a manner similar to DLBCL with doxorubicin remission of the disease, when associated infection is eradi-
(Adriamycin)based regimens. cated with antibiotic therapy.18

Extranodal Marginal Zone Lymphoma of Morphology and Immunophenotype. In most cases,


Mucosa-Associated Lymphoid Tissue the neoplastic population is composed of a mixture of medium-
Definition. Three subtypes of marginal zone lymphomas sized lymphocytes, plasma cells, and occasional large lym-
are recognized: nodal, extranodal (MALT lymphoma), and phoid cells. Medium-sized marginal zone cells with irregular
splenic. In this chapter, we focus on the extranodal variant nuclei and relatively abundant cytoplasm predominate (Figure
derived from marginal zone cells of MALT. In this lymphoma, 37-17, A). Residual reactive germinal centers may be present,
the neoplastic proliferation is usually heterogeneous, encom- which are colonized to variable degrees by the neoplastic cells.
passing small and medium-sized lymphocytes, plasma cells, A characteristic feature of MALT lymphoma is the presence of
and scattered large lymphoid cells. This disease frequently so-called lymphoepithelial lesions, representing the invasion
is associated with autoimmune conditions (e.g., Sjgren of the neoplastic lymphocytes into the glandular epithelium
570 PART V Leukocyte Disorders

TABLE 37-3 Clinicopathologic Features of Selected Plasma Cell Neoplasms


Plasma Cell Disorder Defining Features
Plasma cell myeloma Monoclonal protein in serum or urine
Clonal plasma cells in bone marrow or presence of plasmacytoma
Organ or tissue impairment (CRAB: hypercalcemia, renal insufficiency, anemia, bone lesions)
Plasma cell leukemia Clonal plasma cell population in bone marrow and other features of plasma cell myeloma in conjunction
with peripheral blood involvement: >2 109/L or >20% circulating plasma cells
May present with involvement of spleen, liver, and body cavity fluid including cerebrospinal fluid
Monoclonal gammopathy of Bone marrow plasmacytosis (<10% plasma cells)
undetermined significance Monoclonal gammopathy (<30 g/L)
No lytic bone lesions or organ/tissue impairment
Solitary plasmacytoma of bone Localized bone mass composed of plasma cells
Extraosseous plasmacytoma Localized extraosseous mass composed of plasma cells

(see Figure 37-17, B). This feature is usually absent from may be preceded by an asymptomatic period of monoclonal
reactive lymphoid proliferations associated with autoimmune gammopathy with only mild bone marrow plasmacytosis
processes or infections. (fewer than 10% plasma cells). Approximately 25% of asymp-
The neoplastic cells of marginal zone lymphoma express tomatic patients with clonal serum immunoglobulin progress
CD20, CD19, and monoclonal immunoglobulin chains. CD5 to symptomatic plasma cell myeloma.21 The term monoclonal
and CD10 are absent. CD43 antigen is coexpressed in 30% of gammopathy of undetermined significance (MGUS) is used to
cases and can be a helpful feature in diagnosing marginal zone encompass the entire patient population with clonal serum
lymphoma when there is a significant residual reactive compo- immunoglobulin and only mild marrow plasmacytosis. Plas-
nent. In select cases, the demonstration of clonality by flow macytoma, a localized form of plasma cell neoplasm, may
cytometry or molecular analysis may be necessary to confirm present as a solitary bone lesion or involve an extraosseous or
the diagnosis. extramedullary site, most commonly the nasopharynx, oro-
Antigenic stimulation and interference with the apoptotic pharynx, or sinuses.
pathway play an important role in the pathogenesis of MALT
lymphoma. Approximately 30% of cases show a translocation Morphology and Immunophenotype. Plasma cell
involving apoptosis-inhibitor gene API2 and the MLT gene, the myeloma is characterized by marked bone marrow plasmacy-
t(11;18)(q21;q21).19 tosis. Large aggregates and sheets of plasma cells, frequently
with cytologic atypia, are present and often constitute more
Clinical Features and Prognosis. The gastrointestinal than 30% of marrow cellularity (Figure 37-18, A). Atypical
tract is the most common site for extranodal marginal zone cytologic features seen in plasma cell myeloma include a high
lymphoma. The lung, thyroid, ocular adnexa, and breast are nuclear-to-cytoplasmic ratio, dispersed chromatin pattern, and
other common sites of involvement. In most cases, the disease distinct nucleoli (see Figure 37-18, B). These changes are rarely
is limited to the primary site with occasional involvement of seen in reactive conditions and MGUS. Similarly, in reactive
regional lymph nodes. Bone marrow is less frequently involved plasmacytosis associated with infections and autoimmune dis-
than in other types of indolent lymphoma.20 In cases of gastric orders, plasma cells appear scattered throughout the marrow
MALT lymphoma positive for H. pylori, the antibiotic treatment cavity and form small clusters around the vessels. Large aggre-
of the infection may induce a remission of the associated lym- gates and sheets of plasma cells, which are commonly seen in
phoma. Other cases may benefit from local therapy. plasma cell myeloma, are absent from reactive conditions.
Rarely, patients with plasma cell myeloma show a marked
Plasma Cell Neoplasms increase in circulating plasma cells. The term plasma cell leuke-
Definition. Plasma cell neoplasms are characterized by mia is reserved for cases with more than 20% circulating plasma
a monoclonal proliferation of terminally differentiated B cells or plasma cell counts exceeding 2 109/L. Neoplastic
cells (i.e., plasma cells). These disorders can present as a plasma cells can also infiltrate the spleen, liver, or lymph nodes
localized or disseminated process most commonly involving and involve body cavity fluids (see Figure 37-18, C).
bone marrow and bone. The clinical features and the primary Plasmacytoma represents a localized, mass-forming, mono-
site of involvement define distinct clinicopathologic entities clonal plasma cell proliferation. Neoplastic plasma cells show
(Table 37-3). an immunophenotype similar to that of their normal counter-
Plasma cell myeloma is a multifocal accumulation of parts. At this terminal stage of differentiation, panB-cell
malignant plasma cells in the bone marrow presenting as lytic markers CD19 and CD20 and surface immunoglobulin chains
bone lesions. In most cases, monoclonal immunoglobulin are usually absent. Plasma cells are positive for CD138 (synde-
produced by neoplastic plasma cells is detected in the serum, can 1), high-density CD38 antigen, and monoclonal cytoplas-
urine, or both (monoclonal gammopathy). The overt disease mic immunoglobulins. These monoclonal proteins, in the
CHAPTER 37 Mature Lymphoid Neoplasms 571

form of complete immunoglobulin, most commonly IgG or


IgA or isolated clonal light chains, are secreted by the neoplas-
tic plasma cells and are seen in serum and urine as monoclonal
spikes. In addition, neoplastic plasma cells can show an aber-
rant expression of other antigens typically not expressed by
normal plasma cells, such as CD56 and myeloid markers.

Clinical Features and Prognosis. Plasma cell myeloma


is a disease of older individuals (median age, approximately
70 years).22 Bone pain and pathologic fractures are directly
related to the proliferation of neoplastic plasma cells. These
cells produce factors that cause localized bone destruction
(lytic lesions on radiographic examination) and hypercalce-
mia. Renal insufficiency is triggered by the obstruction or direct
A damage of renal tubules by monoclonal protein. Cytopenias
are related to the replacement of normal trilineage hemato
poiesis by massive plasma cell infiltrates. Depressed normal
immunoglobulin levels confer susceptibility to infections,
which commonly occur in patients with plasma cell myeloma.
High levels of serum immunoglobulins also may interfere with
the coagulation cascade and impair circulation through an
increase in serum viscosity. Tissue deposits of clonal immuno-
globulins, called amyloidosis, may compromise kidney, heart,
and liver function and cause peripheral neuropathies.
Most of the cases show a rapidly progressive course, and
the median survival is 3 years. The prognosis is closely related
to the number of plasma cells in the bone marrow and to
clinical features reflecting overall tumor burden. Patients with
more than 50% bone marrow plasma cells, associated renal
failure, and severe anemia have a shorter survival than
B patients with fewer than 20% plasma cells and preserved renal
function.
Typically, patients with bone and extraosseous plasma
cytomas are younger and respond favorably to local radiation
therapy. Approximately 15% progress to plasma cell myeloma.
Asymptomatic monoclonal gammopathy (MGUS) occurs
in fewer than 5% of individuals older than 70 years.21 As dis-
cussed earlier, 25% of patients with MGUS develop overt
myeloma. That is why all individuals with MGUS should
be monitored closely with repeat measurements of serum
immunoglobulin levels and bone marrow examination.

Diffuse Large B-Cell Lymphoma


Definition. The defining feature of DLBCL is its large cell
size. In contrast to the neoplastic cells in the lymphoprolifera-
tive disorders discussed so far, DLBCL cells are significantly
C
larger than normal lymphocytes. Most show a diffuse histo-
Figure 37-18 Plasma cell myeloma. A, Bone marrow biopsy specimen logic growth pattern and can differ significantly in cytologic
showing large aggregates of plasma cells (hematoxylin and eosin stain, appearance and immunophenotype. DLBCL is one of the most
400). B, Neoplastic plasma cells with cytologic atypia (bone marrow, common lymphomas, accounting for 30% to 40% of all non-
Wright-Giemsa stain, 1000). C, Plasma cells in the pleural fluid of a patient Hodgkin lymphoma cases.9
with plasma cell myeloma (Wright-Giemsa stain, 1000).
Morphology and Immunophenotype. The most
common type, DLBCL not otherwise specified, shows a diffuse
proliferation of large lymphoid cells replacing the normal
lymph node architecture. Cells are at least twice the size of
normal small lymphocytes and show single or multiple
572 PART V Leukocyte Disorders

A
Figure 37-19 Diffuse large B-cell lymphoma with proliferation of large
lymphoid cells. Note the size difference between small lymphocytes (arrow)
and neoplastic B cells (arrowhead) (hematoxylin and eosin stain, 500).

nucleoli and ample cytoplasm (Figure 37-19). In rare cases,


there is a considerable admixture of background histiocytes
and small lymphocytes in addition to the neoplastic large cells.
As in other B-cell lymphomas, panB-cell antigens are
expressed. DLBCL can originate from a variety of stages in
B-cell development; hence the coexpression of other markers
is heterogeneous. CD5, CD10, BCL-6, CD30, and CD138 can
be present (see Table 37-2).

Clinical Features and Prognosis. Although, as with B


most lymphomas, the median age at diagnosis is in the sixties, Figure 37-20 Burkitt lymphoma. A, Starry sky pattern imparted by
DLBCL can be seen in children and young adults. Most com- numerous macrophages with apoptotic debris (hematoxylin and eosin stain,
monly, it presents as a localized disease involving a group of 400). B, Touch preparation showing characteristic cells of Burkitt lym-
lymph nodes. Bone marrow involvement is rare at presentation phoma. Note the deeply basophilic cytoplasm with numerous vacuoles
but can occur later in the course of the disease. DLBCL can also (Wright-Giemsa stain, 1000).
be seen in extranodal sites, including the gastrointestinal tract,
central nervous system, bone, and serous effusions. DLBCL is
an aggressive neoplasm with a proliferation rate frequently prominent starry sky pattern imparted by numerous tingible
exceeding 40%, which makes it more sensitive to multiagent body macrophages (Figure 37-20, A). The macrophages are
chemotherapy. The prognosis depends on a variety of clinical responsible for phagocytosing apoptotic debris, a by-product
parameters, such as patient age, the extent of disease, and the of the extremely high proliferation rate. Lymphoma cells are
site of involvement. The original morphologic subclassification medium-sized with round nuclei, finely clumped chromatin,
of DLBCL was flawed by poor intraobserver and interobserver and small nucleoli. The cytoplasm is deeply basophilic and
reproducibility.23 However, recent results of gene microarray highly vacuolated, a feature best displayed on touch imprints
studies have shown that patients with DLBCL of follicle or other cytologic preparations (see Figure 37-20, B).
center origin have better survival than patients with other The immunophenotype of Burkitt lymphoma reflects ger-
subtypes.24 minal center origin. CD19, CD20, CD10, and BCL-6 antigens
are present. There is surface expression of monoclonal immu-
Burkitt Lymphoma noglobulin light chains. BCL-2 is absent. The hallmark of
Definition. Burkitt lymphoma is characterized by medium- Burkitt lymphoma is a high proliferation rate. Nearly 100% of
sized, highly proliferating lymphoid cells with basophilic Burkitt lymphoma cells are actively proliferating. This feature
vacuolated cytoplasm. The WHO classification lists three vari- is linked to the constitutive expression of MYC gene (cell cycle
ants of this lymphomaendemic (occurring predominantly in gatekeeping gene) secondary to its translocation under the pro-
Africa), sporadic, and immunodeficiency associated. moter of immunoglobulin heavy or light chain genes, t(8;14),
t(2;8), t(8;22). This translocation is pathognomonic for Burkitt
Morphology and Immunophenotype. The lymphoid lymphoma, and its demonstration is required for definitive
proliferation is diffuse, and at low magnification shows a diagnosis.
CHAPTER 37 Mature Lymphoid Neoplasms 573

Clinical Features and Prognosis. The clinical presenta- lymphoid cells with irregular nuclear outlines (cerebriform
tion of Burkitt lymphoma is dependent on the variant nuclei). These cells show a predilection for the epidermis (epi-
(endemic, sporadic, or immunodeficiency associated). The dermotropism) and dermis and may spread to regional lymph
endemic form presents in young children (4 to 7 years of age), nodes. Szary syndrome presents as a disseminated disease
most commonly as a jawbone mass. The sporadic variant, seen with widespread skin involvement (erythroderma), lymphade-
in the United States and Europe, occurs in children and young nopathy, and circulating lymphoma cells (Szary cells with
adults most commonly as an abdominal mass. Gastrointestinal characteristic cerebriform nuclei).
tract and abdominal lymph nodes are often involved; however,
other extranodal sites, such as gonads and breasts, can be a site Morphology and Immunophenotype. In mycosis fun-
of primary disease. Immunodeficiency-associated Burkitt lym- goides, the degree of cutaneous infiltrate is related to the stage
phoma presents most often as nodal disease. Independent of of the disease. Early lesions show patchy or lichenoid infiltrate
the presentation, Burkitt lymphoma commonly involves the of the dermis by small to medium-sized lymphoid cells with
central nervous system, bone marrow, and peripheral blood irregular nuclear outlines (Figure 37-21, A). The colonization
(Burkitt leukemia). Epstein-Barr virus (EBV) is present in a of epidermis by neoplastic lymphocytes is seen (see Figure
proportion of patients. 37-21, B), with small aggregates of lymphoma cells forming
A diagnosis of Burkitt lymphoma is a medical emergency. so-called Pautrier microabscesses. Later in the course of the
Because of its high proliferation rate, the doubling time is disease, cutaneous infiltrates may become more dense and
extremely short. The chemotherapy is significantly different form tumorlike foci. The involvement of regional lymph nodes
from that used for other types of high-grade lymphoma. These and peripheral blood may be present, especially in advanced
highly aggressive treatment regimens take into account the stages of the disease.
high proliferation rate and contribute to high cure rates
for childhood and adult Burkitt lymphoma60% to 90%
depending on the stage of the disease.25,26 Immunodeficiency-
associated Burkitt lymphoma occurs predominantly in HIV-
positive patients; the prognosis is not as favorable as for other
variants.

Mature T-Cell and Natural Killer


Cell Lymphomas
Lymphomas derived from mature T cells and natural killer cells
are much less common than the previously discussed mature
B-cell neoplasms and account for approximately 10% of all
lymphomas. The incidence of the specific subtypes of T-cell
lymphoma shows geographic and ethnic variability. In certain
geographic regions, T-cell malignancies may be more prevalent
than in the United States. Compared with B-cell lymphopro-
liferative disorders, T-cell neoplasms occur more frequently in A
extranodal sites. The most common skin lymphoma, mycosis
fungoides, is of T-cell phenotype.
Although morphology is an important criterion in the diag-
nosis of T-cell lymphomas, a significant morphologic and cyto-
logic variability is seen within specific subtypes. Similarly, the
immunophenotypic features are not as specific as those seen
in B-cell malignancies. Because of these factors, an integration
of morphologic, immunophenotypic, cytogenetic, molecular,
and clinical information, as recommended by the WHO clas-
sification, is crucial in diagnosing T-cell and natural killer cell
malignancies. Until recently, the demonstration of clonality in
T-cell proliferations was limited to molecular methods showing
T-cell receptor gene rearrangements. The development of mul-
tiple antibodies directed against the variable region of the
T-cell receptor allows the determination of T-cell clonality by
flow cytometry.27 B
Figure 37-21 Mycosis fungoides. A, Dermal infiltrate of medium-sized
Mycosis Fungoides and Szary Syndrome lymphoid cells with angulated nuclei (hematoxylin and eosin stain, 400).
Definition. Mycosis fungoides is the most common cuta- B, Neoplastic lymphocytes invading the epidermis (hematoxylin and eosin
neous lymphoma. It is composed of small to medium-sized stain, 400).
574 PART V Leukocyte Disorders

The immunophenotype is similar to that of T lymphocytes


normally present in the skin. The expression of panT-cell
markers CD3, CD5, and CD2 is seen along with CD4 antigen.
An important feature, rarely seen in benign lymphoid infil-
trates, is the absence of CD7 antigen. The T-cell receptor gene
is clonally rearranged.
Szary syndrome is by definition a disseminated disease
with leukemic presentation and skin and lymph node involve-
ment.28,29 The malignant cells are medium-sized with cerebri-
form nuclei. Skin and lymph node involvement is more
pronounced than in early stages of mycosis fungoides and
shows diffuse, monotonous lymphoid infiltrates. The evalua-
tion of peripheral blood involvement based on morphologic
criteria alone is not recommended due to low interobserver
reproducibility. Current hematologic criteria are based on both
morphologic and immunophenotypic evaluation and include
Figure 37-22 Peripheral T-cell lymphoma showing heterogeneous popu-
at least 1000 Szary cells/mm3, a CD4-to-CD8 ratio of more
lation of small, medium-sized, and large lymphoid cells (hematoxylin and
than 10 due to significant numbers of circulating CD4+ Szary
eosin stain, 500).
cells, and the presence of an aberrant immunophenotype.1 The
latter is defined as a significant loss of CD7, CD26, or T-cell
marker(s) on CD4+ T-cells. treatment regimens have been developed specifically for T-cell
lymphomas; aggressive B-cell lymphoma protocols are com-
Clinical Features and Prognosis. The incidence of monly used to treat these disorders.
mycosis fungoides increases with age, and the average age at
presentation is 55 to 60 years. The survival of patients with Anaplastic Large Cell Lymphoma
early-stage disease is excellent, because progression and devel- Definition. Although considerable morphologic variabil-
opment of disseminated lymphoma are very slow.28 In this ity can be seen, the typical case of anaplastic large cell lym-
patient group, 10-year disease-specific survival was reported as phoma is characterized by large atypical cells with pleomorphic
97% to 98%. In most cases, only local treatment is necessary. nuclei and abundant cytoplasm. The expression of CD30
In contrast, Szary syndrome is an aggressive lymphoma with antigen and ALK protein is seen in the majority of cases.
a low (10% to 20%) 5-year survival rate.29
Morphology and Immunophenotype. Numerous mor-
Peripheral T-Cell Lymphoma, Unspecified phologic variants have been described dependent on the pre-
Definition. Peripheral T-cell lymphoma, unspecified, dominant architectural and cytologic features. The lymph node
comprises a morphologically heterogeneous group of lympho- architecture is most often diffusely effaced by malignant lym-
mas with mature T-cell phenotype. phoid cells (Figure 37-23, A). Occasionally, lymph node involve-
ment may be partial with characteristic infiltrates of nodal
Morphology and Immunophenotype. Lymph node sinuses. When significant fibrosis is present, anaplastic large cell
involvement is usually diffuse with a prominent vascular pro- lymphoma may resemble Hodgkin lymphoma. Regardless of
liferation. The cytologic features are variable with medium- the histologic variant, in almost every case, at least a proportion
sized to large cells with atypical and occasionally pleomorphic of cells are large with abundant cytoplasm and pleomorphic,
nuclei (Figure 37-22). Certain variants show a considerable eccentric, kidney-shaped nuclei, so-called hallmark cells. Leuke-
admixture of reactive small lymphocytes, immunoblasts, his- mic involvement is not frequent; however, it may be seen in
tiocytes, and eosinophils. (Bone marrow is frequently involved, the small cell variant. In such cases, the peripheral blood film
but a significant circulating lymphoma component is rarely shows atypical lymphoid cells with indented nuclei reminiscent
seen at presentation.) Most cases are derived from CD4+ T cells of Szary cells (see Figure 37-23, B). In such cases immunophe-
and retain this immunophenotype. Variable loss of panT-cell notyping is instrumental for prompt diagnosis.
antigens, including CD7, is seen. CD30 antigen and, in most cases, ALK protein are defining
immunophenotypic features of this lymphoma. The overex-
Clinical Features and Prognosis. Peripheral T-cell pression of ALK is most often due to t(2;5)(p23;35) between
lymphoma is an aggressive disease occurring predominantly in the ALK and nucleophosmin genes. Alternative partner genes
older adults (average age, 60 years). Generalized lymphade- for ALK translocation have also been identified. Although
nopathy and a variety of constitutional symptoms, such as the cytotoxic T-cell origin of this lymphoma is indisputable,
fever, night sweats, weight loss, and pancytopenia, are present panT-cell markers (CD3, CD7, CD5) are often absent. The
at diagnosis. The 3-year survival rate is reported to be approxi- most commonly expressed T-cell lineagespecific antigens are
mately 40%.30 Biologic features and advanced stage at presenta- CD4 and CD2 and cytotoxic antigens TIA-1, granzyme, and
tion contribute to the dismal prognosis. In addition, few perforin.31 Pan-hematopoietic antigen CD45 is expressed only
CHAPTER 37 Mature Lymphoid Neoplasms 575

A
Figure 37-24 Characteristic popcorn (lymphocytic/histiocytic) cells of
nodular lymphocyte-predominant Hodgkin lymphoma (arrow) (hematoxylin
and eosin stain, 1000).

Clinical Features and Prognosis. Anaplastic large cell


lymphoma is less frequent in adults. However, it is one of the
most common lymphomas in the pediatric population, repre-
senting 10% to 15% of childhood lymphomas. Anaplastic large
cell lymphoma presents as disseminated nodal disease with
constitutional symptoms. Extranodal sites, including skin, can
also be involved. The most important prognostic feature is the
expression of ALK protein. ALK+ disease has a favorable prog-
nosis, whereas ALK disease shows survival rates more compa-
rable to those for peripheral T-cell lymphoma, unspecified.
B
Figure 37-23 A, Anaplastic large cell lymphoma with large pleomorphic Hodgkin Lymphoma
cells (hematoxylin and eosin stain, 1000). B, Peripheral blood involvement Hodgkin lymphoma can be divided into two broad categories:
by small cell variant of anaplastic large cell lymphoma (Wright-Giemsa stain, nodular lymphocyte-predominant Hodgkin lymphoma and
1000). classical Hodgkin lymphoma.1 Although both disorders occur
preferentially in young individuals and share certain morpho-
TABLE 37-4 Immunophenotypic Features of logic characteristics, more recent studies have shown that
Lymphomas Composed of Large Lymphoid Cells they are biologically distinct entities, and they are discussed
separately.
Antigen NLPHL Classical HL DLBCL ALCL
CD30 + +/ + Nodular Lymphocyte-Predominant
CD15 + Hodgkin Lymphoma
CD45 + + +/ Definition. Nodular lymphocyte-predominant Hodgkin
CD20 + +/* + lymphoma is a B-cell neoplasm composed of relatively rare
CD3 +/ neoplastic cells (lymphocytic/histiocytic or popcorn cells)
scattered within nodules of reactive lymphocytes.
ALCL, Anaplastic large cell lymphoma; DLBCL, diffuse large B-cell lymphoma; HL,
Hodgkin lymphoma; NLPHL, nodular lymphocyte-predominant Hodgkin lymphoma.
*If positive, the immunoreactivity is weak and present only in a proportion of neoplastic
Morphology and Immunophenotype. The normal
cells. architecture of a lymph node is replaced by a nodular prolifera-

Other T-cell markers, such as CD2 and CD4, are more often present. tion of small lymphocytes and scattered lymphocytic/histiocytic
or popcorn cells, the latter being the neoplastic cell of nodular
in a proportion of cases (Table 37-4). By flow cytometry, the lymphocyte-predominant Hodgkin lymphoma (Figure 37-24).
coexpression of myeloid markers such as CD13, CD33, and These are large lymphoid cells with abundant cytoplasm and
CD15 is seen in approximately 50% of cases. In cases negative vesicular multilobated nuclei (popcorn nuclei). The nucleoli
for the T-cellassociated antigens and ALK protein, the dem- are inconspicuous.
onstration of clonal T-cell receptor gene rearrangement is The lymphocytic/histiocytic cells are positive for B-cell
pivotal in making the diagnosis. markers, including CD20 antigen (see Table 37-4). These cells
576 PART V Leukocyte Disorders

are of germinal center cell origin32; thus, they express follicular Despite their distinct morphologic characteristics and clini-
marker BCL-6 and immunoglobulin chains. The neoplastic cal features, Reed-Sternberg cells are present in all subtypes of
cells do not show evidence of EBV infection. The surrounding Hodgkin lymphoma. The classic Reed-Sternberg cell is a large
nodular background is composed of CD20+ small B cells and lymphoid cell with a bilobed nucleus or two nuclei with prom-
scattered T lymphocytes. inent eosinophilic nucleoli and abundant cytoplasm (Figure
37-25, A). When encountered in an appropriate background,
Clinical Features and Prognosis. Most patients are Reed-Sternberg cells are pathognomonic for the diagnosis.
males in their thirties and present with localized disease. However, not all neoplastic cells of classical Hodgkin lym-
Mostly peripheral lymph nodes are involved. Mediastinal phoma show the typical morphology of Reed-Sternberg cells.
lymphadenopathy is rare. As for classical Hodgkin lymphoma, Variants of neoplastic cells, including Hodgkin cells, mummi-
the prognosis is excellent, with survival rates of 80% to 90% fied cells, and lacunar cells, are often encountered in a single
when the disease is diagnosed in the early stages.33 lymph node. Hodgkin cells are large mononuclear lymphoid
cells with an oval nucleus, thick nuclear membrane, distinct
Classical Hodgkin Lymphoma eosinophilic nucleolus, and abundant cytoplasm. Mummified
Definition. Classical Hodgkin lymphoma comprises a cells are degenerated or apoptotic cells with a pyknotic nucleus
heterogenous group of lymphoid neoplasms derived from the and condensed cytoplasm. Lacunar cells occur predominantly
germinal center.34 It is characterized by the presence of rela- in the nodular sclerosis variant of classical Hodgkin lymphoma
tively few diagnostic neoplastic cells, Reed-Sternberg cells, in a and are characterized by a lobated nucleus and artifactual
rich reactive background. The incidence of this disease varies retraction of the cytoplasm secondary to formalin fixation.
in different geographic regions. In the United States and Because of this artifact, the cells appear to be situated in a clear
Europe, it is a common form of lymphoma occurring in young space (i.e., lacuna).
adults. In the United States, approximately 7400 new cases are In all subtypes of classical Hodgkin lymphoma, Reed-
diagnosed annually.5 A bimodal age distribution is observed, Sternberg cells and their variants have a similar immunophe-
with incidence peaks between 15 and 34 years and older than notype (see Table 37-4). They are CD30+ in all cases (see Figure
54 years. 37-25, B) and CD15+ in approximately 80% of cases. The
CD15 immunoreactivity may be weak and seen in only a few
Morphology and Immunophenotype. On the basis of malignant cells. The expression of B-cell marker CD20 is weak
architectural features, the composition of the reactive back- to absent. Similarly, CD45 antigen is absent. The frequency of
ground, and relative proportion of neoplastic cells, classical EBV infection depends on the subtype of classical Hodgkin
Hodgkin lymphoma can be divided into four subtypes (Table lymphoma. The background small lymphocytes are predomi-
37-5), as follows: nantly CD4+ T cells.
1. Nodular sclerosis Nodular sclerosis classical Hodgkin lymphoma. The
2. Mixed cellularity defining feature of the nodular sclerosis subtype is the presence
3. Lymphocyte rich of broad collagen bands transecting the lymph node, thicken-
4. Lymphocyte depleted ing of the nodal capsule (see Figure 37-25, C), and lacunar
cells. The background cellularity includes small lymphocytes,
eosinophils, and histiocytes. This is the most common of all
the subtypes of classical Hodgkin lymphoma, accounting for
TABLE 37-5 Morphologic Subtypes of Classical
70% of cases. The frequency of immunohistochemically
Hodgkin Lymphoma
demonstrable EBV infection is lowest in this variant.
Neoplastic Additional Morphologic Mixed cellularity classical Hodgkin lymphoma. In the
Subtype Cells Features mixed cellularity subtype, Reed-Sternberg cells and their vari-
Nodular RS cells; Hodgkin Fibrotic bands; background of ants are scattered among the diffuse background proliferation
sclerosis cells; lacunar small lymphocytes, histiocytes, of small lymphocytes, histiocytes, eosinophils, neutrophils,
cells and eosinophils and plasma cells. Typical Reed-Sternberg cells, mononuclear
Mixed RS cells; Hodgkin Background of small lymphocytes, Hodgkin cells, and mummified cells are seen; however, lacunar
cellularity cells eosinophils, neutrophils, cells are absent. Similarly, fibrotic bands and capsular thicken-
histiocytes, plasma cells; no ing are not seen. Approximately 20% of classical Hodgkin
fibrotic bands lymphomas show this morphology. An association with EBV
Lymphocyte RS cells; Hodgkin Diffuse nodular background of infection is seen in 75% of cases.
rich cells small lymphocytes; no or few Lymphocyte-rich classical Hodgkin lymphoma. Most
eosinophils and neutrophils commonly, in the lymphocyte-rich subtype scattered mono-
Lymphocyte RS cells; Hodgkin Numerous RS cells and Hodgkin nuclear Hodgkin and Reed-Sternberg cells are seen along with
depleted cells lymphoma cells; few a vaguely nodular background of small lymphocytes. Nodules
background lymphocytes represent remnants of mantle zones and residual germinal
centers. Compared with other subtypes of classical Hodgkin
RS, Reed-Sternberg. lymphoma, the background cellularity is less heterogeneous.
CHAPTER 37 Mature Lymphoid Neoplasms 577

A B

C
Figure 37-25 Classical Hodgkin lymphoma. A, Typical Reed-Sternberg cells (arrows) of classical Hodgkin lymphoma with two nuclear lobes and distinct
nucleoli (hematoxylin and eosin stain, 500). B, CD30+ Reed-Sternberg cells and their variants (immunoperoxidase, 500). C, Nodular sclerosis type of clas-
sical Hodgkin lymphoma showing broad fibrotic bands dissecting the lymph node (hematoxylin and eosin stain, 40).

Lymphocyte-depleted classical Hodgkin lymphoma. variant, which often shows mediastinal lymphadenopathy.
The lymphocyte-depleted subtype is an uncommon variant of With the contemporary treatment protocols combining chemo-
classical Hodgkin lymphoma occurring predominantly in therapy and radiotherapy the cure rates are 80% to 90%,
immunodeficient patients. There is a paucity of cells of reactive depending on the stage of the disease, patient age, and clinical
background, and neoplastic Reed-Sternberg cells and their vari- symptoms. The best prognosis is seen in the nodular sclerosis
ants are much more frequent. In most cases, neoplastic cells subtype. Lymphocyte-depleted Hodgkin lymphoma is the most
show evidence of EBV infection. aggressive variant of classical Hodgkin lymphoma, especially
in HIV-positive patients. In this patient group, Hodgkin lym-
Clinical Features and Prognosis. With the exception phoma also may manifest in unusual extranodal sites, includ-
of the lymphocyte-rich variant, which occurs in a slightly older ing bone marrow. Patients with classical Hodgkin lymphoma
population, classical Hodgkin lymphoma is a disease of young treated with a combination of chemotherapy and radiotherapy
adults with a peak incidence at 15 to 35 years. Mostly peripheral are at high risk of developing secondary malignancies, includ-
lymph nodes are involved except in the nodular sclerosis ing lung and breast carcinomas and acute leukemia.

SUMMARY
Histologic components of normal lymph nodes include the cortex, Lymphomas are neoplastic lesions of the lymphoid system arising
paracortex, medullary cords, and sinuses. These are structural at specific stages of lymphoid differentiation.
and functional compartments from which reactive hyperplasias Modern lymphoma classification incorporates morphologic, immu-
and malignant disorders originate. nophenotypic, molecular, and clinical characteristics.
578 PART V Leukocyte Disorders

Lymphomas are broadly divided into lesions derived from precur- B-cell and T-cell lymphomas and Hodgkin lymphoma involve
sor (immature) and mature lymphoid cells, and B-cell and T-cell mainly lymph nodes; the involvement of extranodal sites and bone
neoplasms. marrow or peripheral blood occur with varying frequency.
The most common mature B-cell neoplasms are follicular lym- In general, lymphomas occur in elderly individuals; however, spe-
phoma and DLBCL. cific subtypes such as Hodgkin lymphoma show a predilection for
The T-cell neoplasms most common in the United States and younger age groups.
Europe are peripheral T-cell lymphoma, unspecified, and anaplas- The prognosis depends on lymphoma subtype. Indolent lympho-
tic large cell lymphoma. mas show a protracted course but are largely incurable with
In CLL, the peripheral blood and bone marrow display smudge current chemotherapeutic regimens. In contrast, aggressive lym-
cells and small lymphoid cells with coarse chromatin, inconspicu- phomas have a more rapidly progressive course, and the cure
ous nucleoli, and scant cytoplasm. rates are higher.
In HCL, small B lymphocytes have abundant cytoplasm and fine
cytoplasmic projections. Now that you have completed this chapter, go back and
Hodgkin lymphoma has been shown to be of B-cell origin. read again the case study at the beginning and respond
Hodgkin lymphoma is subclassified based on morphologic and to the questions presented.
immunophenotypic features.

R E V I E W Q UESTIONS
1. In most cases, the diagnosis of lymphoma relies on all of 6. What is the major morphologic difference between
the following except: Hodgkin lymphoma and other B-cell lymphomas?
a. Microscopic examination of affected lymph nodes a. The extent of the lymph node involvement
b. Immunophenotyping using immunohistochemistry or b. The presence of numerous reactive lymphocytes and
flow cytometry only a few malignant cells in Hodgkin lymphoma
c. Molecular or cytogenetic analysis c. The presence of numerous tingible body macrophages
d. Peripheral blood examination and a complete blood in Hodgkin lymphoma
count d. The preservation of normal lymph node architecture
in Hodgkin lymphoma
2. The most common lymphoma occurring in young
adults is: 7. Which morphologic diagnosis has to be confirmed with
a. Follicular lymphoma molecular studies demonstrating the presence of t(8;14)?
b. DLBCL a. Mantle cell lymphoma
c. Hodgkin lymphoma b. Burkitt lymphoma
d. Mycosis fungoides c. Follicular lymphoma
d. Szary syndrome
3. In a normal lymph node, the medulla includes
predominantly: 8. What is the function of the germinal center?
a. T cells a. Generation of B cells producing immunoglobulins
b. B cells with the highest affinity for a particular antigen
c. Tingible body macrophages through the process of somatic mutation
d. Plasma cells b. Production of plasma cells that secrete specific
immunoglobulins following antigenic stimulation
4. The t(11;14) is the defining feature of: c. T-cell maturation following their education in the
a. Follicular lymphoma thymus
b. Hodgkin lymphoma d. Generation of dendritic cells with unique antigen-
c. CLL processing capabilities
d. Mantle cell lymphoma
9. Marked paracortical expansion is most often associated
5. The immunophenotype of mycosis fungoides is: with:
a. The normal T-cell immunophenotype a. Rheumatoid arthritis
b. An abnormal T-cell immunophenotype with b. Syphilis
expression of CD4 and loss of CD7 antigen c. Dermatopathic lymphadenopathy
c. A mix of CD4+ and CD8+ T cells d. Follicular lymphoma
d. An abnormal T-cell immunophenotype with
expression of CD8 and loss of CD7 antigen
CHAPTER 37 Mature Lymphoid Neoplasms 579

10. MGUS is best described as: c. The presence of significant bone marrow
a. The presence of monoclonal immunoglobulin in plasmacytosis in a patient with only a few clinical
serum with only mild bone marrow plasmacytosis symptoms of plasma cell myeloma
(fewer than 10% plasma cells) d. The presence of monoclonal immunoglobulin in a
b. The presence of monoclonal serum or urine patient with a solitary mass composed of plasma cells
immunoglobulin with significant bone marrow
plasmacytosis

REFERENCES
1. Swerdlow SH, Campo E, Harris NL, et al, editors: WHO clas- 20. Thieblemont C, Berger F, Dumontet C, et al: Mucosa-
sification of tumours of haematopoietic and lymphoid tissues, associated lymphoid tissue lymphoma is a disseminated
ed 4, Lyon, France, 2008, IARC Press. disease in one third of 158 patients analyzed. Blood 95:802-
2. MacLennan IC: Germinal centers. Annu Rev Immunol 12:117- 806, 2000.
139, 1994. 21. Kyle RA, Therneau TM, Rajkumar SV, et al: Long-term
3. Manser T: Textbook germinal centers? J Immunol 172:3369- follow-up of 241 patients with monoclonal gammopathy of
3375, 2004. undetermined significance: the original Mayo Clinic series 25
4. Herman GE, Chlipala E, Bochenski G, et al: Zinc formalin years later. Mayo Clin Proc 79:859-866, 2004.
fixative for automated tissue processing. J Histotechnol 11:85- 22. Kyle RA, Rajkumar SV: Multiple myeloma. N Engl J Med
89, 1988. 351:1860-1873, 2004.
5. Greenlee RT, Hill-Harmon MB, Murray T, et al: Cancer statis- 23. Harris NL, Jaffe ES, Stein H, et al: A revised European-
tics, 2001. CA Cancer J Clin 51:15-36, 2001. American classification of lymphoid neoplasms: a proposal
6. Filipovich AH, Mathur A, Kamat D, et al: Primary immuno- from the International Lymphoma Study Group. Blood
deficiencies: genetic risk factors for lymphoma. Cancer Res 84:1361-1392, 1994.
52(19 suppl):5465s-5467s, 1992. 24. Alizadeh AA, Eisen MB, Davis RE, et al: Distinct types of
7. Mueller N: Overview of the epidemiology of malignancy in diffuse large B-cell lymphoma identified by gene expression
immune deficiency. J Acquir Immune Defic Syndr 21(suppl profiling. Nature 403:503-511, 2000.
1):S5-S10, 1999. 25. Blum KA, Lozanski G, Byrd JC: Adult Burkitt leukemia and
8. Fisher SF, Fisher RI: The epidemiology of non-Hodgkins lym- lymphoma. Blood 104:3009-3020, 2004.
phoma. Oncogene 23:6524-6534, 2004. 26. Cairo MS, Sposto R, Perkins SL, et al: Burkitts and Burkitt-like
9. The Non-Hodgkins Lymphoma Classification Project: A clin- lymphoma in children and adolescents: a review of the Chil-
ical evaluation of the International Lymphoma Study Group drens Cancer Group experience. Br J Haematol 120:660-670,
Classification of non-Hodgkins lymphoma. Blood 89:3909- 2003.
3918, 1997. 27. Clark DM, Boylston AW, Hall PA, et al: Antibodies to T cell
10. Pileri SA, Sabattini E, Agostinelli C, et al: Histopathology of antigen receptor beta chain families detect monoclonal T cell
B-cell chronic lymphocytic leukemia. Hematol Oncol Clin proliferation. Lancet 2:835-837, 1986.
North Am 18:807-826, 2004. 28. Fink-Puches R, Zenahlik P, Bck B, et al: Primary cutaneous
11. Stilgenbauer S, Bullinger L, Lichter P, et al: Review: genetics lymphomas: applicability of current classification schemes
of chronic lymphocytic leukemia: genomic aberrations and (European Organization for Research and Treatment of
VH gene mutation status in pathogenesis and clinical course. Cancer, World Health Organization) based on clinicopatho-
Leukemia 16:993-1007, 2002. logic features observed in a large group of patients. Blood
12. Shanafelt TD, Ghia P, Lanasa MC, et al: Monoclonal B-cell 99:800-805, 2002.
lymphocytosis (MBL): biology, natural history and clinical 29. Vonderheid EC, Bernengo MG, Burg G, et al: Update on
management. Leukemia 24:512-520, 2010. erythrodermic cutaneous T-cell lymphoma: report of the
13. Guidelines on the diagnosis and management of chronic International Society for Cutaneous Lymphomas. J Am Acad
lymphocytic leukaemia. Br J Haematol 125:294-317, 2004. Dermatol 46:95-106, 2002.
14. Dohner H, Fischer K, Bentz M, et al: p53 gene deletion pre- 30. Escalon MP, Liu NS, Yang Y, et al: Prognostic factors and
dicts for poor survival and non-response to therapy with treatment of patients with T-cell non-Hodgkin lymphoma.
purine analogs in chronic B-cell leukemias. Blood 85:1580- Cancer 103:2091-2098, 2005.
1589, 1995. 31. Foss HD, Anagnostopoulos I, Araujo I, et al: Anaplastic large-
15. Swerdlow SH, Williams ME: From centrocytic to mantle cell cell lymphomas of T-cell and null-cell phenotype express
lymphoma: a clinicopathologic and molecular review of cytotoxic molecules. Blood 88:4005-4011, 1996.
three decades. Hum Pathol 33:7-20, 2002. 32. Braeuninger A, Kuppers R, Strickler JG, et al: Hodgkin and
16. Horsman DE, Gascoyne RD, Coupland RW, et al: Com Reed-Sternberg cells in lymphocyte predominant Hodgkin
parison of cytogenetic analysis, southern analysis, and poly- disease represent clonal populations of germinal center
merase chain reaction for the detection of t(14;18) in derived tumor B cells. Proc Natl Acad Sci U S A 94:9337-9342,
follicular lymphoma. Am J Clin Pathol 103:472-478, 1995. 1997.
17. Thieblemont C, Berger F, Coiffier B: Mucosa-associated lym- 33. Diehl V, Sextro M, Franklin J, et al: Clinical presentation,
phoid tissue lymphomas. Curr Opin Oncol 7:415-420, 1995. course, and prognostic factors in lymphocyte-predominant
18. Parsonnet J, Isaacson P: Bacterial infection and MALT lym- Hodgkins disease and lymphocyte-rich classical Hodgkins
phoma. N Engl J Med 350:213-215, 2004. disease: report from the European Task Force on Lymphoma
19. Dierlamm J, Baens M, Wlodarska I, et al: The apoptosis inhib- Project on Lymphocyte-Predominant Hodgkins Disease.
itor gene AP12 and a novel 18q gene, MLT, are recurrently J Clin Oncol 17:776-783, 1999.
rearranged in the t(11;18)(q21;q21) associated with mucosa- 34. Thomas RK, Re D, Wolf J, et al: Part I: Hodgkins lymphoma
associated lymphoid tissue lymphomas. Blood 93:3601-3609, molecular biology of Hodgkin and Reed-Sternberg cells.
1999. Lancet Oncol 5:11-18, 2004.
PART VI Hematology in Selected Populations

38 Pediatric and Geriatric Hematology Linda H. Goossen

OUTLINE OBJECTIVES
Pediatric Hematology After completion of this chapter, the reader will be able to:
Prenatal Hematopoiesis
Hematopoiesis of the 1. Describe the major differences in normal complete 8. Explain the clinical significance of anemia in the
Newborn blood count values, reticulocytes, and nucleated red elderly.
Pediatric Developmental blood cells (NRBCs) in preterm newborns, full-term 9. Name the two most common anemias seen in
Stages newborns, infants, children, adults, and elderly the elderly and their common causes in this age
Gestational Age and Birth adults. group.
Weight 2. Explain the cause of physiologic anemia of infancy 10. List other anemias affecting elderly individuals.
Red Blood Cell Values at and the time frame in which it is expected. 11. Compare the frequency of acute lymphoblastic
Birth 3. Describe normal RBC morphology in neonates. leukemias and chronic lymphocytic leukemias in
White Blood Cell Values 4. Compare the RBC survival in preterm and full-term children and the elderly.
in the Newborn
infants with that in adults. 12. Compare the hemostatic systems of newborns, chil-
Platelet Values in the
5. Recognize and list factors affecting sample collection dren, adults, and the elderly, including the risk of
Newborn
Neonatal Hemostasis that can have an impact on the interpretation of bleeding and thrombosis.
Geriatric Hematology hematology values in newborns. 13. Name malignancies of hematologic cells that are
Aging and Hematopoiesis 6. Compare and contrast the morphology of lympho- more common in the elderly than in other age
Assessment of cytes in children and in adults and indicate reasons groups.
Hematologic for differences.
Parameters in Healthy 7. Describe the general association between age and
Elderly Adults hemoglobin levels in the elderly.
Anemia and the Elderly
Hematologic Neoplasia in
Older Individuals
Geriatric Hemostasis

CASE STUDY
After studying this chapter, the reader should be able to respond to the following case study:

A full-term newborn infant in no apparent distress had a CBC WBC differential: 50% neutrophils and 50% lymphocytes
performed as part of a panel of testing for infants born to RBC morphology: Macrocytic with slight to moderate polychromasia.
mothers who received no prenatal care. There were 7 NRBCs/100 WBCs.
Results were as follows:
1. Are the results for hemoglobin and hematocrit within the
Patient Results Reference Range limits expected for a newborn?
WBCs (109/L) 18.0 9-37 2. What is the significance of the elevated MCV, NRBCs, and
RBCs (1012/L) 5.28 4.10-6.10 polychromasia?
Hb (g/dL) 18.5 16.5-21.5 3. Comment on the WBC and differential count.
Hct (%) 55.5 48-68

580
CHAPTER 38 Pediatric and Geriatric Hematology 581

H
ematologic values are fairly stable throughout adult time of birth, the bone marrow is fully active and extremely
life, but significant differences exist in the pediatric cellular, with all hematopoietic cell lineages undergoing cel-
and, to some extent, in the geriatric populations. This lular differentiation and amplification. In addition to the
chapter focuses on the more significant differences. mature cells in fetal blood, there are significant numbers of
circulating progenitor cells in cord blood.7,8
In a full-term infant, hepatic hematopoiesis has ceased
PEDIATRIC HEMATOLOGY
except in widely scattered small foci that become inactive soon
Children are not merely small adults. The newborn infant, after birth.1-6 Postembryonic extramedullary hematopoiesis is
older child, and adult exhibit profound hematologic differ- abnormal in a full-term infant. In a premature infant, foci of
ences from one another. Because children mature at different hematopoiesis are frequently seen in the liver and occasionally
rates, it is inappropriate to use adult reference ranges for the observed in the spleen, lymph nodes, or thymus.1-10
assessment of pediatric blood values. Historically, pediatric
reference values were inferentially established from adult data Pediatric Developmental Stages
because of the limitations in attaining analyzable data. Pediat- Pediatric hematologic values change markedly in the first
ric procedures required large blood draws and tedious meth- weeks and months of life. As a result, many variables influence
odologies and lacked standardization. The implementation the interpretation of what might be considered normal values
of child-friendly phlebotomy techniques and micropediatric at the time of birth. It is important to provide age-appropriate
procedures has revolutionized laboratory testing. Pediatric pediatric hematology values that extend from neonatal life
hematology has emerged as a specialized science with age- through adolescence. The pediatric population can be catego-
specific reference ranges that correlate with the hematopoietic, rized with reference to three different developmental stages:
immunologic, and chemical changes in a developing child. (1) the neonatal period, which represents the first 4 weeks of
Dramatic changes occur in the blood and bone marrow of life; (2) infancy, which incorporates the first year of life; and
the newborn infant during the first hours and days after birth, (3) childhood, which spans age 1 to puberty (age 8 to12 years).
and there are rapid fluctuations in the quantities of all hema- Preterm, low-birth-weight infants are more apt to develop
tologic elements. Significant hematologic differences are seen health problems than are other newborns. Since the 1990s, the
between term and preterm infants and among newborns, rising quality of medical care in neonatal intensive care units
infants, young children, and older children. This chapter has improved markedly the survival of smaller infants born
reviews neonatal hematopoiesis, which is discussed in detail at younger gestational ages with less mature hematopoietic
in Chapter 7, as a prerequisite to understanding the changes in systems.
pediatric hematologic reference ranges, morphologic features,
and age-specific physiology. Gestational Age and Birth Weight
Hematologic values obtained from full-term infants generally
Prenatal Hematopoiesis do not apply to preterm infants, and laboratory values for low-
Hematopoiesis, the formation and development of blood birth-weight preterm infants differ from ranges for extremely-
cells from stem cells, begins in the first weeks of embryonic low-birth-weight micropreemies (24 to 26 weeks gestation).11
development and proceeds systematically through three phases A full-term infant is defined as an infant who has completed
of development: mesoblastic (yolk sac), hepatic (liver), and 37 to 42 weeks of gestation. Infants born before 37 weeks
myeloid (bone marrow). The first cells produced in the devel- gestation are referred to as premature or preterm, whereas infants
oping embryo are primitive erythroblasts formed in the yolk delivered after 42 weeks are considered postterm.11,12 Infants can
sac. These cells are particularly interesting because they do be subcategorized further by birth weight as (1) appropriate
not develop into mature erythrocytes. They are erythropoietin size for gestational age; (2) small for gestational age, including
insensitive and have the ability to differentiate into other cell low-birth-weight infants (2500 g or less); (3) very-low-birth-
lines upon exposure to appropriate growth factors.1-6 weight infants (1500g or less); (4) extremely-low-birth-weight
By the second month of gestation, hematopoiesis ceases in micropreemies (500g or less); and (5) large for gestational age
the yolk sac, and the liver becomes the center for hematopoi- (more than 4000g).11
esis, reaching its peak activity during the third and fourth
gestational months. Leukocytes of each cell type systematically Red Blood Cell Values at Birth
make their appearance. In week 9 of gestation, lymphocytes Neonatal hematologic values are affected by the gestational age
can be detected in the region of the thymus. They are subse- of the infant, the age in hours after delivery, the presence of
quently found in the spleen and lymph nodes. During the illness, and the level of support required. Other important
fourth and fifth gestational months, the bone marrow emerges variables to be considered when evaluating laboratory data
as a major site of blood cell production, and it becomes the include site of sampling and technique (capillary versus venous
primary site by birth (see Chapter 7).1-6 puncture, warm or unwarmed extremity), timing of sampling,
and conditions such as the course of labor and the treatment
Hematopoiesis of the Newborn of the umbilical vessels.11,13,14 The presence of fetal hemoglobin
Hematopoietically active bone marrow is referred to as red (Hb F), bilirubin, and lipids in newborns can also interfere
marrow, as opposed to inactive yellow (fatty) marrow. At the with hematology laboratory testing.5-17 As with all laboratory
582 PART VI Hematology in Selected Populations

Figure 38-1 Peripheral blood film for a normal newborn demonstrating Figure 38-2 Peripheral blood film for a premature infant showing a
a normal lymphocyte, macrocytes, polychromasia, and one nucleated red normal lymphocyte, four nucleated red blood cells, and increased polychro-
blood cell (1000). masia (500).

testing, each laboratory should establish reference intervals than a week in immature infants. The average number of
based on its instrumentation, methods, and patient population NRBCs ranges from 3 to 10 per 100 white blood cells (WBCs)
(see Chapter 5). in a normal full-term infant to 25 NRBCs per 100 WBCs in a
premature infant. The presence of NRBCs for more than 5 days
Red Blood Cell Count suggests hemolysis, hypoxic stress, or acute infection.*
Refer to the inside front cover of this book for red blood cell The erythrocytes of newborns show additional morphologic
(RBC) reference ranges. The RBC count increases during the differences. The number of biconcave discs relative to stomato-
first 24 hours of life, remains at this plateau for about 2 weeks, cytes is reduced in neonates (43% discs, 40% stomatocytes)
and then slowly declines. This polycythemia of the newborn18 compared with adults (78% discs, 18% stomatocytes).27 In
may be explained by in utero hypoxia, which becomes more addition, increased numbers of pitted cells, echinocytes, sphe-
pronounced as the fetus grows. Hypoxia, the trigger for rocytes, and other abnormally shaped erythrocytes are seen in
increased secretion of erythropoietin, stimulates erythropoie- neonates. The number of these dysmorphic cells is even
sis.19 At birth, the physiologic environment changes, and the higher in premature infants. Zipursky etal found 40% discs,
fetus makes the transition from its placenta-dependent oxygen- 30% stomatocytes, and 27% additional poikilocytes in prema-
ation to the increased tissue oxygenation of the lungs. This ture infants.27
increased oxygen tension suppresses erythropoietin produc-
tion, which is followed by a decrease in RBC and hemoglobin Reticulocyte Count
production. Studies show that erythropoietin levels before An apparent reticulocytosis exists during gestation, decreasing
birth are equal to or greater than adult levels with a gradual from 90% reticulocytes at 12 weeks gestation, to 15% at 6
drop to near zero a few weeks after birth.20-23 This decline cor- months gestation, and ultimately to 4% to 6% at birth. Reticu-
responds to the physiologic anemia seen at 5 to 8 weeks of life, locytosis persists for about 3 days after birth, then declines
with the RBCs reaching their lowest count at 7 weeks of age abruptly to 0.8% reticulocytes on postnatal day 4 to 7. At 2
and hemoglobin reaching its lowest concentration at 9 weeks months, the number of reticulocytes increases slightly, fol-
of age.19 lowed by a slight decline from 3 months to 2 years, when adult
levels of 0.5% to 1.5% are attained. The reticulocyte count of
Erythrocyte Morphology of the Neonate. Early nor- premature infants is typically higher than that of term infants;
moblasts are megaloblastic, hypochromic, and irregularly however, the count can vary dramatically depending upon how
shaped. During hepatic hematopoiesis, normoblasts are ill the newborn is. Significant polychromasia seen on a Wright-
smaller than the megaloblasts of the yolk sac but are still mac- stained blood film is indicative of postnatal reticulocytosis
rocytic. Erythrocytes remains macrocytic from the first 11 (Figure 38-2).
weeks of gestation until day 5 of postnatal life (Figure 38-1).*
The macrocytic RBC morphology gradually changes to the Hemoglobin
characteristic normocytic, normochromic morphology. Ortho- Full-Term Infants. Hemoglobin synthesis results from
chromic normoblasts frequently are observed in the full-term an orderly evolution of a series of embryonic, fetal, and adult
infant on the first day of life but disappear within postnatal hemoglobins. At birth Hb F constitutes 70% to 80% of the total
days 3 to 5. These nucleated RBCs (NRBCs) may persist longer
*References 1, 2, 6, 13, 14, 20-24, 26.

*References 1, 2, 13, 14, 20, 21, 24, 25. References 1, 2, 6, 13, 14, 20-24, 26.
CHAPTER 38 Pediatric and Geriatric Hematology 583

TABLE 38-1 Hematologic Values for Very-Low-Birth- concentration decreases to approximately 11g/dL, erythropoi-
Weight Infants During the First 6 Weeks of Life etic activity increases until it reaches its adult levels by 14 years
of age.11,18,35-40 Also contributing to the physiologic anemia is
AGE OF INFANT (DAYS)
the shortened life span of the fetal RBC. Studies of chromium-
Hematologic Value 3 12-14 24-26 40-42
labeled newborn RBCs estimate a survival time of 60 to 70 days
Hemoglobin (g/dL) 15.6 14.4 12.4 10.6 with correction for the elution rate of chromium from newborn
Hematocrit (%) 47 44 39 33 cells.1 The life span of RBCs in premature infants is about 35
Red blood cells (1012/L) 4.2 4.1 3.8 3.4 to 50 days.1 The more immature the infant, the greater the
Reticulocytes (%) 7.1 1.7 1.5 1.8 degree of reduction.1,21-23,26,41 The reasons for the shortened life
Platelets (109/L) 203.5 318 338 357 span of the erythrocytes are unclear. This physiologic anemia
White blood cells (109/L) 9.5 12.3 10.4 9.1 is not known to be associated with any abnormalities in the
infant. Hemodilution related to the increased blood volume
Modified from Obladen M, Diepold K, Maier RF, etal: Venous and arterial hematologic
that accompanies the rapid weight gain seen in the first few
profiles of very low birth weight infants, Pediatrics 106:707-711, 2000.
months of life is not thought to play a key role in the anemia.1
The hemoglobin levels of premature infants are typically
hemoglobin.28 Hb F declines from 90% to 95% at 30 weeks 1g/dL or more below the values of full-term infants. Thereaf-
gestation to approximately 7% at 12 weeks after birth and ter, a gradual recovery occurs, which results in values approxi-
stabilizes at 3.2 2.1% at 16 to 20 weeks after birth.29 The mating those of normal full-term infants by about 1 year of
switch from Hb F to Hb A is genetically controlled and deter- age.* Very-low-birth-weight infants (less than 1500g) show a
mined by gestational age; it does not appear to be influenced progressive decline in hemoglobin, RBC count, mean cell
by the age at which birth occurs.30 Chapter 10 provides an volume (MCV), mean cell hemoglobin (MCH), and mean cell
in-depth discussion of the ontogeny, structure, and types of hemoglobin concentration (MCHC), and have a slower recov-
hemoglobin. ery than other preterm and term infants.
The concentration of hemoglobin fluctuates dramatically in
the weeks and months after birth as a result of physiologic Hematocrit
changes, and various factors must be considered when analyz- The average hematocrit (Hct) at birth for full-term infants is
ing pediatric hematologic values. The site of sampling, gesta- 53% (range, 48% to 68%).42 Frequently, newborns with
tional age, and time interval between delivery and clamping of increased hematocrits, especially values greater than 65%,
the umbilical cord can influence the hemoglobin level in experience hyperviscosity of the blood. This can cause prob-
newborn infants.* In addition, there are significant differences lems in producing a high-quality peripheral blood film.
between capillary and venous blood hemoglobin levels. Capil- The hematocrit usually increases approximately 5% during
lary samples in newborns generally have a higher hemoglobin the first 48 postnatal hours; this is followed by a slow linear
concentration than venous samples, which can be attributed decline to 46% to 62% at 2 weeks and 32% to 51% between
to circulatory factors. Racial differences must also be consid- the second and fourth months. Normal adult values of
ered when evaluating hemoglobin levels in children. African 47% for males and 42% for females are achieved during
American children have hemoglobin levels averaging 0.5g/dL adolescence.
lower than those in white children.31 Very-low-birth-weight preterm infants are frequently anemic
The average hemoglobin for a full-term infant at birth is at birth (see Table 38-1). Many require transfusions or eryth-
16.5 to 21.5g/dL; levels less than 14g/dL are considered ropoietin injections or both.
abnormal.20,21 The average hemoglobin value for a preterm
infant who is small for gestational age is 17.1g/dL, lower than Red Blood Cell Indices
that for a full-term infant; hemoglobin values less than 13.7g/ The RBC indices and RBC distribution width (RDW) provide
dL are considered abnormal in preterm infants.21 a means for assessing and defining anemia (see Chapter 18).

Physiologic Anemia of the Neonate. The hemoglobin Mean Cell Volume. The erythrocytes of newborn infants
concentration of term infants decreases during the first 5 to 8 are markedly macrocytic at birth. The average MCV for full-
weeks of life, a condition known as physiologic anemia of infancy. term infants is 110 15fL; however, a sharp decrease occurs
Infants born prematurely also experience a decrease in hemo- during the first 24 hours of life. The MCV continues to decrease
globin concentration, which is termed physiologic anemia of to 90 12fL in 3 to 4 months.18,37 The more premature the
prematurity.1,32-34 Along with a decrease in hemoglobin, there is infant, the higher the MCV. A newborn with an MCV of less
a reduction in the number of RBCs, a decrease in the reticulo- than 94fL should be evaluated for -thalassemia or iron
cyte percentages (Table 38-1), and undetectable levels of eryth- deficiency.1,43
ropoietin associated with the transition from the placenta to
the lungs as a source of oxygen. When the hemoglobin Mean Cell Hemoglobin. MCH is 30 to 42pg in healthy
neonates and 27 to 41pg in premature infants.18,37
*References 1, 2, 6, 9-11, 13.

References 11, 13, 14, 20-22, 24, 25. *References 15, 18, 23, 26, 35, 36, 40.
584 PART VI Hematology in Selected Populations

Mean Cell Hemoglobin Concentration. The average birth and reaching adult values at 6 months.53 Both serum fer-
MCHC is the same for full-term infants, premature infants, and ritin and serum iron are high at birth, rise during the first
adults, approximately 33g/dL. month, drop to their lowest level between 6 months and 4
years of age, and remain low throughout childhood.54-56 Con-
Red Blood Cell Distribution Width. The RDW is mark- sideration of these differences is important when interpreting
edly elevated in newborns, with a range of 14.2% to 19.9% the hematology laboratory results for infants and children.
first 30 days of life. After that it gradually decreases, and reaches
normal adult levels by 6 months of age.25 White Blood Cell Values in the Newborn
Fluctuations in the number of WBCs are common at all ages
Anemia in Infants and Children but are greatest in infants (Table 38-2). Leukocytosis is typical
Nutritional deficiencies in infants and children can result in at birth for full-term and preterm infants, with a wide range of
iron deficiency anemia and, rarely, in megaloblastic anemia normal.57 There is an excess of segmented neutrophils and
(see Chapter 20), particularly in low-birth-weight and prema- bands, and an occasional metamyelocyte, with no evidence of
ture infants. These anemias are associated with abnormal psy- disease. The neutrophilic leukocyte count rises within the first
chomotor development; however, they can easily be treated 12 hours following birth, decreases between 1 month and 1
with dietary fortification.44-48 year, then gradually increases to stabilize at 4.4 109/L at about
4 years of age.57
Iron Deficiency Anemia. Iron deficiency anemia is the
most common pediatric hematologic disorder and the most Neutrophilic Leukocytes
frequent cause of anemia in childood.49 The occurrence of iron Refer to Table 38-2 and the inside front cover for the leukocyte
deficiency anemia in infants has decreased in the United States reference ranges for full-term infants. Term and premature
due to iron fortification of infant formula and increased rates infants show a greater absolute neutrophil count than that
of breastfeeding.50 However, the prevalence is still 2% in tod- found in older children, who characteristically maintain a pre-
dlers 1 to 2 years of age and 3% in children 3 to 5 years of dominance of lymphocytes. Band forms are also higher for the
age51 and is related to early introduction and excessive intake first 3 to 4 days after birth (Table 38-3). Newborn females have
of whole cows milk.44,52 Refer to Chapter 19 for an in-depth neutrophil counts averaging 2000 cells/mcL higher than those
discussion of iron deficiency anemia. of males; neonates whose mothers have undergone labor have
higher counts than neonates delivered by cesarean section with
Ancillary Tests for Anemia in Infants and Children. no preceding maternal labor.58,59 There is some evidence that
The differential diagnosis of anemia in infants and children absolute neutrophil counts are lower in healthy black children
relies on a variety of ancillary tests. The reference ranges for a than in white children.60
number of these tests differ from those for adults. Haptoglobin
levels are so low as to be undetectable in neonates, which Premature Infants. At birth, preterm infants exhibit a
makes it unreliable as a marker of infant hemolysis.53 Transfer- left shift, with promyelocytes and myelocytes frequently
rin levels are also lower in neonates, increasing rapidly after observed. The trend to lymphocyte predominance occurs later

TABLE 38-2 Normal Leukocyte Counts*


TOTAL LEUKOCYTES NEUTROPHILS LYMPHOCYTES MONOCYTES EOSINOPHILS
Age Mean Range Mean Range % Mean Range % Mean % Mean %

Birth 4.0 2.0-6.0 4.2 2.0-7.3 0.6 0.1
12hr 11.0 7.8-14.5 4.2 2.0-7.3 0.6 0.1
24hr 9.0 7.0-12.0 4.2 2.0-7.3 0.6 0.1
1-4wk 3.6 1.8-5.4 5.6 2.9-9.1 0.7 0.2
6mo 11.9 6.0-17.5 3.8 1.0-8.5 32 7.3 4.0-13.5 61 0.6 5 0.3 3
1yr 11.4 6.0-17.5 3.5 1.5-8.5 31 7.0 4.0-10.5 61 0.6 5 0.3 3
2yr 10.6 6.0-17.0 3.5 1.5-8.5 33 6.3 3.0-9.5 59 0.5 5 0.3 3
4yr 9.1 5.5-15.5 3.8 1.5-8.5 42 4.5 2.0-8.0 50 0.5 5 0.3 3
6yr 8.5 5.0-14.5 4.3 1.5-8.0 51 3.5 1.5-7.0 42 0.4 5 0.2 3
8yr 8.3 4.5-13.5 4.4 1.5-8.0 53 3.3 1.5-6.8 39 0.4 4 0.2 2
10yr 8.1 4.5-13.5 4.4 1.8-8.0 54 3.1 1.5-6.5 38 0.4 4 0.2 2
16yr 7.8 4.5-13.0 4.4 1.8-8.0 57 2.8 1.2-5.2 35 0.4 5 0.2 3
21yr 7.4 4.5-11.0 4.4 1.8-7.7 59 2.5 1.0-4.8 34 0.3 4 0.2 3

From Cairo MS, Bracho F: White blood cells. In Rudolph CD, Rudolph AM, Hostetter MK, etal, editors: Rudolphs pediatrics, ed 21, New York, 2003, McGraw-Hill, p 1548.
*Numbers of leukocytes are 109/L (or thousands per microliter), ranges are estimates of 95% confidence limits, and percentages refer to differential counts.

Neutrophils include band cells at all ages and a small number of metamyelocytes and myelocytes in the first few days of life.

Dashes indicate insufficient data for a reliable estimate.


CHAPTER 38 Pediatric and Geriatric Hematology 585

TABLE 38-3 Neutrophil and Band Counts for Newborns


during the First 2 Days of Life*
Absolute Neutrophil Absolute Band
Age Count ( 109/L) Count ( 109/L)
Birth 3.5-6.0 1.3
12hr 8.0-15.0 1.3
24hr 7.0-13.0 1.3
36hr 5.0-9.0 0.7
48hr 3.5-5.2 0.7

Modified from Luchtman-Jones L, Schwartz AL, Wilson DB: Hematologic problems


in the fetus and neonate. In Martin RJ, Fanaroff AA, Walsh MC, editors: Fanaroff
and Martins neonatal-perinatal medicine, ed 8, Philadelphia, 2006, Mosby Elsevier,
p 1310.
*Normal values were obtained from the assessment of 3100 separate white blood cell
counts obtained from 965 infants; 513 counts were from infants considered to be Figure 38-3 Peripheral blood film for a normal newborn showing a benign
completely normal at the time the count was obtained and for the preceding and
subsequent 48 hours. There was no difference in the normal ranges when values were lymphocyte (1000).
stratified by infant birth weight or gestational age.

than in full-term infants. The neutrophil counts in premature


infants are similar to or slightly lower than the neutrophil
counts in full-term infants during the first 5 days of life;
however, the count gradually declines to 2.5 109/L (1.1 to
6.0 109/L) at 4 weeks.61 There is no significant difference in
the neutrophil count of infants by birth weight or gestational
age; however, very-low-birth-weight infants have a significantly
lower limit (1 109/L) compared with larger infants.61-63

Neutropenia. Neutropenia is defined as a reduction in the


number of circulating neutrophils to less than 1.5 109/L.
Neutropenia accompanied by bands and metamyelocytes is
often associated with infection, particularly in preterm neo-
nates. Neutropenia represents a decrease in neutrophil produc-
tion or an increase in consumption.64 Figure 38-4 Peripheral blood film for a normal newborn showing a benign
lymphocyte with visible nucleoli (1000).
Neutrophilia. Neutrophilia refers to an increase in the
number of neutrophils to greater than 8 109/L. Morphologic
changes associated with infection include Dhle bodies, be similar in appearance to the malignant cells seen in child-
vacuoles, and toxic granulation.65 hood acute lymphoblastic leukemia (ALL), these benign cells
lack the asynchronous or aberrant antigen expression seen in
Eosinophils and Basophils ALL and thus can be differentiated from the lymphocytes of
The percentages of eosinophils and basophils remain consis- infant ALL by immunophenotyping (see Chapter 33).71,72
tent throughout infancy and childhood. Refer to the inside
front cover of this book for reference ranges. Monocytes
The mean monocyte count of neonates is higher than adult
Lymphocytes values. At birth the average proportion of monocytes is 6%.
Lymphocytes constitute about 30% of the leukocytes at birth During infancy and childhood, an average of 5% is maintained
and increase to 60% at 4 to 6 months. They decrease to 50% except in the second and third weeks, when the proportion
by 4 years, to 40% by 6 years, and to 30% by 8 years.13,14,20,24 increases to around 9%. The count reaches adult levels at 3 to
Benign immature B cells (hematogones), although predomi- 5 months.
nantly found in the bone marrow, can sometimes be seen in
the peripheral blood of newborns. These lymphocytes are pri- Neonatal Hematologic Response to Infection. Sepsis
marily mid-stage B cells66,67 and are frequently referred to as in neonates is a common cause of morbidity, particularly in
baby or kiddie lymphocytes. They vary in size from 10 to premature and low-birth-weight infants.73-77 Defective B-cell
20m, have scant cytoplasm and condensed but homogeneous response against polysaccharide agents as well as abnormal
nuclear chromatin, and may have small, indistinct nucleoli cytokine release by neutrophils and monocytes has been
(Figures 38-3 and 38-4).68-70 Although these lymphocytes may implicated.78-81 Because of the transient neutrophilia that
586 PART VI Hematology in Selected Populations

TABLE 38-4 Normal Platelet Counts for Full-Term and 6 months.89 In the fibrinolytic system, levels of plasminogen
Preterm Infants and 2-antiplasmin are similar to adult levels at birth, whereas
levels of tissue plasminogen activator are low and levels of
Platelet Count ( 109/L;
plasminogen activator inhibitor (PAI) are increased (Table
Age mean 1 SD)
38-5).90 The hemostatic components are not only changing in
Preterm infants, 27-31wk 275.0 60.0 concentration over the first few weeks to months of life, but
Preterm infants, 32-36wk 290.0 70.0 their values are also dependent upon the gestational age of the
Term infants 310.0 68.0 child, and premature infants have different values at birth than
Normal child/adult 300.0 50.0 term infants (Table 38-6).

Adapted from Oski FA, Naiman JL: Normal blood values in the newborn period. Bleeding and Thrombosis. The reference ranges for the
In Hematologic problems in the newborn, Philadelphia, 1982, WB Saunders.
SD, Standard deviation. prothrombin time and partial thromboplastin time for neo-
nates extend higher than those for adults; however, these
values normalize by 6 months (see Table 38-5). The risk of
occurs during the first 24 hours after birth, followed by a rapid bleeding is not increased in a healthy newborn despite the
decline, the neutrophil count is not a satisfactory index of decreased levels of the vitamin Kdependent coagulation
infection in the newborn.82 Newborns with bacterial infections factors, which is primarily related to the reduced levels of the
frequently have normal or below normal neutrophil counts physiologic anticoagulants protein C and protein S.91
with a shift to the left. Thus, many practitioners depend upon The risk of thrombosis is considerably less in neonates and
the band count and its derived immature-to-total neutrophil children than in adults. However, two age-related peaks in
ratio as an indicator of sepsis in neonates,83 although CD64, frequency occurthe first in the neonatal period and the
C-reactive protein, and procalcitonin levels have been sug- second in postpuberty adolescence.92 The presence of central
gested as more sensitive markers of sepsis in infants and venous catheters, cancer, and chemotherapy are the most
children.83,84 common risk factors in both of these groups.93-96 Hemorrhagic
and thrombotic disorders are discussed in Chapters 41 and 42,
Platelet Values in the Newborn respectively.
The platelet count usually ranges from 150 to 400 109/L for
full-term and preterm infants, comparable to adult values
GERIATRIC HEMATOLOGY
(Table 38-4).85,86 Platelet counts generally increase in both term
and preterm infants in the first few months of life, as evidenced The life expectancy and quality of life of the elderly have
by increased mean platelet volume the first month of life. improved dramatically in recent decades. Americans are living
Thrombocytopenia of fewer than 100 109 platelets/L may be longer than ever before. Life expectancies at both age 65 and
seen in high-risk infants with sepsis or respiratory distress and age 85 have increased. Under current conditions, people who
neonates with trisomy syndromes, and investigation should be survive to age 65 can expect to live an average of 18.6 more
undertaken for underlying pathology.87,88 Platelets of a newborn yearsalmost 7 years longer than people aged 65 in 1900. The
infant show great variation in size and shape. Neonatal throm- life expectancy of people who survive to age 85 today is 7.2
bocytopenia is discussed in Chapter 43. years for women and 6.1 years for men.97 The U.S. Census
Bureau projects that in 2020 about 1 in 6 Americans will be
Neonatal Hemostasis elderly (age 65 and over) and 6.5 million persons will be age
The physiology of the hemostatic system in infants and chil- 85 and older.98 Global aging is also occurring at a record-
dren is different from that in adults (see Chapter 40). The breaking rate. The World Health Organization (WHO) reports
vitamin Kdependent coagulation factors (factors II, VII, IX, that by 2050, one fifth of the global population will be adults
and X) are at about 30% of adult values at birth; they reach aged 65 years and older, and the number of centenarians will
adult values after 2 to 6 months, although the mean values reach 1.1 million.97
remain lower in children than in adults. Levels of factor XI, Disease and disabilities are not a function of age, although
factor XII, prekallikrein, and high-molecular-weight kininogen age is a risk factor for many diseases. However, with the increase
are between 35% and 55% of adult values at birth, reaching in the aging population, the incidence of age-related health
adult values after 4 to 6 months. In contrast, the levels of conditions also is likely to increase. Thus, the care of the elderly
fibrinogen, factor VIII, and von Willebrand factor are similar has become a growing concern as the life expectancy of the
to adult values throughout childhood.89,90 Factor V decreases population continues to increase.
during childhood, with lower levels during the teen years as The elderly can be roughly divided into three age categories:
compared with adults. The physiologic anticoagulants and (1) the young-old, aged 65 to 74; (2) the old-old, aged 74
inhibitors of coagulationprotein C, protein S, antithrombin, to 84; and (3) the very old, aged 85 and older.99 The 85 and
and a disintegrin-like and metalloprotease domain with throm- older age group is the fastest growing segment of the elderly
bospondin type 1 motifs 13 (ADAMTS 13)are reduced to population.
about 30% to 40% at birth. Antithrombin reaches adult values Geriatric medicine is a rapidly growing branch of medicine.
by 3 months, whereas protein C does not normalize until after The use of inappropriate reference values may lead to
CHAPTER 38 Pediatric and Geriatric Hematology 587

TABLE 38-5 Reference Intervals for Coagulation Tests in the Healthy Full-Term Infant during the First 6 Months of Life
Day 1 Day 5 Day 30 Day 90 Day 180 Adult
Screening Tests
PT (Sec) 10.1-15.9 10.0-15.3 10.0-14.3 10.0-14.2 10.7-13.9 10.8-13.9
PT (INR) 0.53-1.62 0.53-1.48 0.53-1.26 0.53-1.26 0.61-1.17 0.64-1.17
PTT (Sec) 31.3-54.3 25.4-59.8 32.0-55.2 29.0-50.1 28.1-42.9 26.6-40.3
TCT (Sec) 19.0-28.3 18.0-29.2 19.4-29.2 20.5-29.7 19.8-31.2 19.7-30.3

Factor Assays
Fibrinogen (mg/dL) 167-399 162-462 162-378 150-379 150-387 156-400
II (%) 26-70 33-93 34-102 45-105 60-116 70-146
V (%) 34-108 45-145 62-134 48-132 55-127 62-150
VII (%) 28-104 35-143 42-138 39-143 47-127 67-143
VIII (%) 50-178 50-154 50-157 50-125 50-109 50-149
VWF (%) 50-287 50-254 50-246 50-206 50-197 50-158
IX (%) 15-91 15-91 21-81 21-113 36-136 55-163
X (%) 12-68 19-79 31-87 35-107 38-118 70-152
XI (%) 10-66 23-87 27-79 41-97 49-134 67-127
XII (%) 13-93 11-83 17-81 25-109 39-115 52-164
PK (Fletcher, %) 18-69 20-76 23-91 41-105 56-116 62-162
HMWK (Fitzgerald, %) 6-102 16-132 33-121 30-146 36-128 50-136
XIIIa (%) 27-131 44-144 39-147 36-172 46-162 55-155
XIIIb (%) 30-122 32-180 39-173 48-184 20-170 57-137

Control Proteins
Antithrombin (%) 39-87 41-93 48-108 73-121 84-124 79-131
2-Macroglobulin (%) 95-183 98-198 106-194 126-226 149-233 52-120
C1 lnhibitor (%) 36-108 60-120 47-131 71-159 89-193 71-131
1-Antitrypsin (%) 49-137 49-129 36-88 42-102 47-107 55-131
Heparin Cofactor II (%) 10-93 0-96 10-87 10-146 50-190 66-126
Protein C (%) 17-53 20-64 21-65 28-80 37-81 64-128
Protein S (%) 12-60 22-78 33-93 54-118 55-119 60-124

Fibrinolytic Proteins
Plasminogen (%) 125-265 141-293 126-270 174-322 221-381 248-424
Tissue Plasminogen Activator (ng/mL) 5.0-18.9 4.0-10.0 1.0-6.0 1.0-5.0 1.0-6.0 1.4-8.4
2-Antiplasmin (%) 55-115 70-130 76-124 76-140 83-139 68-136
Plasminogen Activator Inhibitor 1 (%) 20-151 0-81 0-88 10-153 60-130 0-110

From Andrew M, Paes B, Johnston M: Development of the hemostatic system in the neonate and young infant, Am J Pediatr Hematol Oncol 12(1):95-104, 1990.
HMWK, High-molecular-weight kininogen; INR, international normalized ratio; PK, prekallikrein; PT, prothrombin time; PTT, partial thromboplastin time; TCT, thrombin clotting
time; VWF, von Willebrand factor.

unnecessary testing and investigations or, more importantly, musculoskeletal, pulmonary, and bone marrow reserve.
result in failure to detect a critical underlying disease. The Certain cells lose their ability to divide (e.g., nervous tissue,
growing concern about the interpretation of hematologic data muscles), whereas others, such as bone marrow and the
in reference to age is due partly to the tremendous heterogene- gastrointestinal mucosa, remain mitotic. Marrow cellularity
ity in the aging process and partly to the difficulty in separating begins at 80% to 100% in infancy and decreases to about
the effects of age from the effects of occult diseases that accom- 50% after 30 years, followed by a decline to 30% after age
pany aging.100 This section focuses on hematologic changes in 65.101-103 These changes may be due to a reduction in the
the elderly and discusses hematologic reference ranges for volume of cancellous (spongy) bone along with an increase in
various geriatric age groups as well as hematopathologic condi- fat, rather than to a decrease in hematopoietic tissue.103 Telo-
tions seen in the geriatric population. mere shortening, which determines the number of divisions a
cell undergoes, has not been definitively related to age-related
Aging and Hematopoiesis bone marrow stem cell exhaustion in humans104; however, it
The aging process is associated with the functional decline is speculated to be associated with hematopoietic stem cell
of several organ systems, such as cardiovascular, renal, differentiation.105
588 PART VI Hematology in Selected Populations

TABLE 38-6 Reference Intervals for Coagulation Tests in the Healthy Premature Infant of 30 to 36 Weeks Gestation
During the First 6 Months of Life
Day 1 Day 5 Day 30 Day 90 Day 180 Adult
Screening Tests
PT (Sec) 10.6-16.2 10.0-15.3 10.0-13.6 10.0-14.6 10.0-15.0 10.8-13.9
PT (INR) 0.61-1.70 0.53-1.48 0.53-1.11 0.53-1.32 0.05-1.48 0.64-1.17
PTT (Sec) 27.5-79.4 26.9-74.1 26.9-62.5 28.3-50.7 27.2-53.3 26.6-40.3
TCT (Sec) 19.2-30.4 18.8-29.4 18.8-29.9 19.4-30.8 18.9-31.5 19.7-30.3

Factor Assays
Fibrinogen (mg/dL) 150-373 160-418 150-414 150-352 150-360 156-400
II (%) 20-77 29-85 36-95 30-106 51-123 70-146
V (%) 41-144 46-154 48-156 59-139 58-146 62-150
VII (%) 21-113 30-138 21-145 31-143 47-151 67-143
VIII (%) 50-213 53-205 50-199 58-188 50-187 50-149
VWF (%) 78-210 72-219 66-216 75-184 54-158 50-158
IX (%) 19-65 14-74 13-80 25-93 50-120 55-163
X (%) 11-71 19-83 20-92 35-99 35-119 70-152
XI (%) 8-52 13-69 15-71 25-93 46-110 67-127
XII (%) 10-66 9-69 11-75 15-107 22-142 52-164
PK (Fletcher, %) 9-57 25-75 31-87 37-121 40-116 62-162
HMWK (Fitzgerald, %) 9-89 24-100 16-112 32-124 41-125 50-136
XIIIa (%) 32-108 57-145 39-147 71-155 65-161 55-155
XIIIb (%) 35-127 68-158 57-157 75-167 67-163 57-137

From Andrew M, Paes B, Milner R, etal: Development of the human coagulation system in the healthy premature infant, Blood 72(5):1651-1657, 1988.
HMWK, High-molecular-weight kininogen; INR, international normalized ratio; PK, prekallikrein; PT, prothrombin time; PTT, partial thromboplastin time; TCT, thrombin clotting
time; VWF, von Willebrand factor.

Assessment of Hematologic Parameters unchanged. Elderly women have hemoglobin concentrations


in Healthy Elderly Adults ranging from 11.7 to 13.8g/dL. Males characteristically have
In 1930, Wintrobe published hematologic normal ranges that higher hemoglobin levels than females, owing to the stimulat-
are still in use today. These were derived from healthy young ing effect of androgens on erythropoiesis; however, the differ-
adults, mostly medical students and nurses. What constitutes ence narrows as androgen levels decrease in elderly men and
normal for elderly patients is a matter of considerable debate. estrogen levels decrease in older women.109,110 Characteristi-
There is controversy concerning the assignment of geriatric cally, the lowest hemoglobin levels are found in the oldest
age-specific reference ranges, especially because aging is often patients (Table 38-7).
accompanied by physiologic changes, and the prevalence of
disease increases markedly. The baseline values for the elderly Leukocytes
are the same reference values used for healthy adults. Proper In the absence of any underlying pathologic condition, there
interpretation of hematologic data, however, requires a com- are no statistically significant differences between the total
plete understanding of the association between disease and leukocyte count and WBC differential for the young-old and
older age. The next section highlights hematologic reference old-old and those for middle-aged adults.108 Some investiga-
ranges for healthy elderly individuals with and without evi- tors, however, have reported a lower leukocyte count, 3.1 to
dence of underlying disease. 8.5 109/L, in individuals older than age 65, owing primarily
to a decrease in the lymphocyte count. Others have reported a
Red Blood Cells decrease in the lymphocyte and the neutrophil counts in
Most RBC parameters (e.g., RBC count and indices and RDW) women, but not in men, older than age 50.109
for normal healthy elderly do not show significant deviations
from those for younger adults. There is a gradual decline in Immune Response in the Elderly. Infectious diseases
hemoglobin starting at middle age, with the mean level decreas- are an important cause of morbidity and mortality in the
ing by about 1g/dL during the sixth through eighth decades.106 elderly. Aged adults are more susceptible to infection, take
Men older than 60 years have average hemoglobin levels of longer to recover from infection, and are often less responsive
12.4 to 15.3g/dL, whereas men aged 96 to 106 years have a to vaccination.111 The adverse changes that occur in the func-
mean hemoglobin level of 12.4g/dL.107,108 The hemoglobin tion of the immune system with age are called immunosenes-
levels in women may increase slightly with age or remain cence.112 Although all components of immunity are affected, T
CHAPTER 38 Pediatric and Geriatric Hematology 589

TABLE 38-7 Hematologic Values in Ambulatory Healthy older individuals.111 The TLRs play a crucial role in the immune
Adults* response.121

84-98yr 30-50yr
Platelets
12
Red blood cells ( 10 /L) The platelet count is not significantly changed with age. There
Males 4.8 0.4 5.1 0.2 have been reports of increased levels of -thromboglobulin
Females 4.5 0.3 4.6 0.3 and platelet factor 4 in the -granules and increased platelet
Hemoglobin (g/dL) phospholipid content.122,123 Thrombocytopenia may be drug
Males 14.8 1.1 15.6 0.7 induced or secondary to marrow infiltration of metastatic
Females 13.6 1.0 14.0 0.8 cancer, lymphoma, or leukemia. Thrombocytosis can be cate-
Hematocrit (%) gorized into primary and secondary. Essential thrombocytosis
Males 43.8 3.3 45.3 2.2 is a myeloproliferative neoplasm characterized by sustained
Females 40.7 2.9 41.6 2.3 proliferation of megakaryocytes resulting in platelet counts of
Mean cell volume (fL) 450 109/L or greater (see Chapter 34).124 Primary thrombo-
Males 91.3 5.4 87.8 2.8 cytosis can also be seen in chronic myelogenous leukemia.
Females 90.5 4.1 90.5 5.0 Secondary thrombocytosis, or reactive thrombocytosis, is asso-
Mean cell hemoglobin (pg) ciated with infections, rheumatoid arthritis, chronic inflamma-
Males 31.0 2.0 30.1 1.6 tory bowel disease, iron deficiency anemia, sickle cell anemia,
Females 30.2 1.2 30.3 2.1 and splenectomy.125-127
Mean cell hemoglobin concentration (g/dL)
Males 33.7 1.5 34.2 1.5 Anemia and the Elderly
Females 33.4 1.2 33.5 1.5 Anemia is common in the elderly and its prevalence increases
White blood cells (109/L) 7.6 0.5 8.8 0.4 with age; however, anemia should not be viewed as an inevi-
Platelets (109/L) 277 2.1 361.0 38.0 table consequence of aging.128,129 Although anemia in the
Neutrophils (109/L) 4.5 0.3 5.9 0.3 elderly is typically mild, it has been associated with substantial
Lymphocytes (109/L) 1.9 0.3 1.9 0.8 morbidity and mortality.130,131 WHO defines anemia as hemo-
globin less than 13g/dL in males and less than 12g/dL in
Adapted from Zauber NP, Zauber AG: Hematologic data of healthy very old people,
females. Using the WHO definition of anemia, the Third
JAMA 357:2181-2184, 1987; and Chatta GS, Lipschitz DA: Aging of the hematopoi-
etic system. In Hazzard WR, Blass JP, Ettinger WH, etal: Principles of geriatric medi- National Health and Nutrition Examination Survey (NHANES
cine and gerontology, ed 4, New York, 1999, McGraw-Hill, p 894. III), a national study that samples clinical specimens, found
*Mean values 1 standard deviation.
the prevalence of anemia in the United States in individuals
over the age of 65 to be 11.0% in men and 10.2% in women.132
cells appear to be the most susceptible.113 The thymus disap- However, studies of the prevalence of anemia in the elderly
pears by early middle age, and adults must then depend on show great variability in the definitions of anemia used as
their T-lymphocyte pool in the secondary tissues to mediate well as differences in sample sizes, patient populations
T celldependent immune responses.114,115 The number of studied, countries in which the study was conducted, and study
naive T cells decreases in the elderly, which increases the design. Thus, estimates of the prevalence of anemia vary from
dependence on memory T cells. T cells of the elderly have 2.9% to 61% in elderly men and from 3.3% to 41% in elderly
impaired responsiveness to mitogens and antigens as a result women.133
of decreased expression of costimulator CD28. There is also an The factors contributing to anemia include a decrease
alteration of T-cell signaling with aging.116-118 B-lymphocyte in bone marrow function, a decline in physical activity,
function depends on T-cell interaction. Thus, the decreased nutritional deficiencies, cardiovascular disease, and chronic
ability to generate antibody responses, especially to primary inflammatory disorders. Anemia of chronic inflammation, iron
antigens, may be the result of T-cell changes rather than intrin- deficiency anemia, and unexplained anemia are the most
sic defects in B lymphocytes.114,115,119,120 common anemias in the elderly.
Many neutrophil functions are decreased in the elderly, Ineffective erythropoiesis and hypoproliferation may also
including chemotaxis, phagocytosis of microorganisms, and be seen in the elderly. Ineffective erythropoiesis is associated
generation of superoxide. Studies indicate that these defects with vitamin B12 or folate deficiency, sideroblastic anemia, and
may be associated with changes to the cell membrane and/or thalassemia in the elderly. Hypoproliferative anemia often
to receptor signalling.111 occurs secondary to iron deficiency, vitamin B12 or folate
deficiency, renal failure, hypothyroidism, chronic inflamma-
Monocytes and Macrophages tion, or endocrine disease.134 To a lesser extent, the elderly are
The aging process does not significantly affect the number of prone to anemias such as aplastic anemia, hemolytic anemia,
monocytes. Information on the effects of aging on monocyte myelophthisic anemia, and anemia due to protein-calorie
and macrophage function is limited and often conflicting.111 malnutrition.
Recent studies provide evidence for defects in the toll-like The initial laboratory evaluation of anemia should include
receptor (TLR) function in monocytes and macrophages in a complete blood count (CBC), a reticulocyte count, peripheral
590 PART VI Hematology in Selected Populations

TABLE 38-8 Classification of Geriatric Anemia meats containing heme iron. Iron deficiency in the elderly
Based on Mean Cell Volume (MCV) and Red Cell Distribution most frequently results from conditions leading to chronic
Width (RDW) gastrointestinal blood loss, including long-term use of nonste-
roidal antiinflammatory medications, gastritis, peptic ulcer
MCV RDW Anemia
disease, gastroesophageal reflux disease with esophagitis, colon
Normal Normal Anemia of chronic inflammation (some) cancer, and angiodysplasia.146-148 It also may be due to poor
Hemorrhagic anemia diet in an elderly individual who has lost the taste or desire for
Leukemia-associated anemia food or is unable to prepare nutritious meals. Chapter 19 dis-
Normal High Early iron deficiency anemia cusses iron disorders in detail.
Mixed deficiency anemia (e.g., vitamin B12 and iron)
Sideroblastic anemia Ineffective Erythropoiesis
Low Normal Anemia of chronic inflammation (some) Ineffective erythropoiesis has been attributed not only to
Low High Iron deficiency anemia maturation disorders such as vitamin B12 and folic acid defi-
High Normal Anemia associated with myelodysplastic syndrome ciency, but also to sideroblastic anemia and thalassemia.
High High Vitamin B12 deficiency anemia Sideroblastic anemias are characterized by impaired heme
Folate deficiency anemia synthesis, and abnormal globin synthesis occurs in the tha
Hemolytic anemia lassemias (see Chapter 27). Megaloblastic anemia results
from defective synthesis of deoxyribonucleic acid (DNA) (see
Chapter 20) with compromised cell division, but normal cyto-
plasmic development (i.e., asynchrony).149-151 These megalo-
blood film review, and chemistry panel, along with other diag- blastic cells are more prone to destruction in the bone marrow,
nostic tools including iron studies (with ferritin), vitamin B12 which results in ineffective erythropoiesis. Two causes of
and folate levels, and free erythrocyte protoporphyrin (Table megaloblastic anemia are deficiency of vitamin B12 and folate
38-8). In addition, assessment for signs of gastrointestinal deficiency.
blood loss, hemolysis, nutritional deficiencies, malignancy,
chronic infection, renal or hepatic disease, or other chronic Vitamin B12 Deficiency. Vitamin B12 (cobalamin) defi-
disease can provide important information for the evaluation ciency causes a megaloblastic disorder in 5% to 10% of the
of anemia in the elderly. elderly.134 It may be difficult to detect, because anemia is
present in only about 60% of patients.152 Neurologic complica-
Anemia of Chronic Inflammation tions are found in 75% to 90% of individuals with clinically
Anemia of chronic inflammation, also known as anemia of apparent vitamin B12 deficiency.153,154 In the absence of anemia,
chronic disease, frequently occurs with inflammatory disorders neurologic symptoms may be the only indication. Even when
(e.g., rheumatoid arthritis, renal failure, liver disease), chronic anemia is present, it does not always manifest with the classic
infections, bedsores, collagen vascular disease, protein malnu- macrocytic and megaloblastic picture but may be normocytic
trition, endocrine disorders, vitamin C deficiency, and neo or microcytic.
plastic disorders.134-142 This hypoproliferative anemia is the Vitamin B12 deficiency in the elderly has been attributed to
most common form of anemia in the hospitalized geriatric inadequate intestinal absorption of food-bound vitamin B12
population.143 The severity of the anemia generally correlates and pernicious anemia rather than inadequate intake.155
with the severity of the underlying disease.144 An impaired Many elderly individuals have atrophic gastritis resulting in
erythropoietin-dependent erythropoiesis triggered by proin- decreased gastric production of acid. In this condition there
flammatory cytokines, impaired iron mobilization, and short- is low vitamin B12 absorption because protein-bound vitamin
ened RBC survival is involved in the pathogenesis of anemia B12 is not dissociated and cannot bind to intrinsic factor. In
of chronic inflammation.145 Chapter 19 discusses anemia of addition, the loss of gastric acid can result in bacterial over-
chronic inflammation. growth, particularly with Helicobacter pylori, which also inter-
feres with vitamin B12 absorption.156,157 Inadequate vitamin B12
Iron Deficiency Anemia absorption in the elderly has also been reported in other
Iron deficiency anemia is common in the elderly. Iron defi- infrequent conditions such as small bowel disorder, gastric
ciency affects not only erythrocytes, but also the metabolic resection, pancreatic insufficiency, resection of the terminal
pathways of iron-dependent tissue enzymes.139 Hemoglobin ileum, blind loop syndrome, and tropical sprue.158 Pernicious
synthesis is reduced, and even a minimal decrease can cause anemia develops slowly and insidiously in patients when
profound functional disabilities in an elderly patient. The autoimmune antibodies to intrinsic factor or to parietal cells
serum iron level decreases progressively with each decade of destroy their parietal cells so that they are left without intrinsic
life, particularly in females. Nevertheless, healthy elderly adults factor.159 Given the high risk of vitamin B12 deficiency in the
usually have normal serum iron levels. aged and the risks of the condition, some authors have
Iron deficiency anemia in the elderly is rarely due to dietary proposed that all elderly adults be screened periodically for
deficiency in industrialized nations because of the prevalence vitamin B12 deficiency.160-162 Chapter 20 discusses megaloblas-
of iron fortification of grains as well as a diet that includes tic anemias.
CHAPTER 38 Pediatric and Geriatric Hematology 591

Folate Deficiency. A second megaloblastic anemia that Myeloproliferative Neoplasms. Myeloproliferative neo-
may be seen in the elderly results from folate deficiency. plasms are monoclonal proliferations of hematopoietic stem
In contrast to vitamin B12 deficiency, folic acid deficiency cells with overaccumulation of RBCs, WBCs, or platelets in
usually develops from inadequate dietary intake, because various combinations. Myeloproliferative disorders include
the body stores little folate.110,163 However, the incidence of chronic myelogenous leukemia, polycythemia vera, essential
low serum and RBC folate levels has declined in all age thrombocythemia, primary myelofibrosis, myeloid neoplasms
groups, including the elderly, since countries began to with eosinophilia and tyrosine kinase mutations, chronic eosi
fortify their grains with folic acid in the 1990s.164 Alcoholic nophilic leukemia not otherwise classified, mast cell disease,
elderly patients are particularly prone to folic acid deficiency, chronic neutrophilic leukemia, and unclassifiable myelopro
because alcohol interferes with folate absorption, enterohe- liferative neoplasms. The incidence of myeloproliferative neo-
patic recycling, metabolism, breakdown, and excretion (see plasms increases from 6.3 per 100,000 for individuals aged 60
Chapter 20).165,166 to 69, to 11.6 per 100,000 for those aged 70 to 79, and to 13.8
per 100,000 for those 80 years of age and older.169 Chapter 34
Hemolytic Anemia discusses the myeloproliferative neoplasms.
Hemolytic anemias are characterized by a shortened RBC sur-
vival time. There are three major types of hemolytic anemias: Leukemia
(1) those caused by immunologic mechanisms; (2) those Leukemia is a neoplastic disease characterized by a malignant
due to intrinsic defects, such as an RBC membrane defect, proliferation of hematopoietic stem cells in the bone marrow,
abnormal hemoglobins, or RBC enzyme defects; and (3) those peripheral blood, and often other organs. Leukemia is broadly
resulting from extrinsic factors, such as mechanical or lytic classified on the basis of the cell type involved (lymphoid
processes.135,167 The elderly are at risk for drug-induced anemia or myeloid) and the stage of maturity of the leukemic cells
because they may take multiple medications. Drug-induced (acute or chronic). Although the overall incidence of leukemia
hemolytic anemia has been associated with high doses of anti- has decreased in the past 5 decades, there has been a dispro-
biotics (penicillin, chloramphenicol, cephalosporins), several portionately greater incidence in leukemia in the elderly
nonsteroidal antiinflammatory drugs, quinidine, phenacetin, (Table 38-9). The peaks in leukemia incidence seem to occur
and others. Hemolytic anemias in the elderly also can result in the very young (age 1 to 4 years) and the very old (age
from collagen vascular diseases, infections, and chronic lym- 70 or older, especially men). Chronic myelogenous leukemia
phocytic leukemia. The hemolytic anemias are discussed in (see Chapter 34) and chronic lymphocytic leukemia (see
Chapters 24 and 25. Chapter 37) show the most dramatic age-related increase in
incidence.
Approximately 13% of patients with acute leukemia have
Hematologic Neoplasia in Older Individuals had previous hematologic disorders, such as myelodysplastic
Although hematologic malignancies may occur at any age, syndrome, hypoplastic bone marrow, polycythemia, or myelo-
certain disorders are common in those older than 50 years. A fibrosis. Geriatric patients are more likely than younger patients
brief overview of these is included in this chapter, with refer- to die due to the toxicity of, or resistance to, chemotherapy.169-171
ences to more detailed discussions.
Geriatric Hemostasis
Chronic Myeloid Neoplasms Age-related changes occur in the vascular and hemostatic
The 2008 WHO classification of adult chronic myeloid systems, including alterations in platelets, coagulation, and
neoplasms includes four broad categories: myelodysplastic
syndrome, myeloproliferative neoplasms, myelodysplastic
syndrome/myeloproliferative neoplasm overlap, and myeloid TABLE 38-9 SEER Incidence of Leukemia in the Elderly
neoplasms associated with eosinophilia and abnormalities of in the United States per 100,000 (2002-2007)*
platelet-derived growth factor receptor , platelet-derived
Age at Diagnosis All Leukemias ALL CLL AML CML
growth factor receptor , or fibroblast growth factor receptor 1.
The chronic myeloid neoplasms show increased incidence in 65-69yr 34.9 1.4 16.0 9.5 4.1
the elderly. 70-74yr 47.3 1.5 20.9 14.0 5.6
75-79yr 62.7 1.6 27.5 18.7 7.5
Myelodysplastic Syndrome. Myelodysplastic syn- 80-84yr 74.4 1.5 30.7 23.0 9.2
drome represents a heterogeneous group of clonal bone 85yr 79.8 1.7 33.2 21.2 9.2
marrow disorders that may affect multiple cell lineages. Myelo-
dysplasia is the most common hematologic malignancy in the Adapted from Altekruse SF, Kosary CL, Krapcho M, etal, editors: SEER cancer statis-
tics review, 1975-2007, Bethesda, Md, 2010, National Cancer Institute. Based on
elderly.168 The incidence of myelodysplastic syndrome increases November 2009 SEER data submission, posted to the SEER website in 2010. Available
from 8.9 cases per 100,000 for individuals aged 60 to 69 to at: http://seer.cancer.gov/csr/1975_2007/. Accessed July 10, 2010.
26.2 per 100,000 for those aged 70 to 79, and is 47.6 per ALL, Acute lymphoblastic leukemia; CLL, chronic lymphocytic leukemia; AML, acute
myeloid leukemia; CML, chronic myelogenous leukemia.
100,000 for those aged 80 and older.169 See Chapter 35 for a *Surveillance, Epidemiology, and End Results (SEER) rates are age adjusted to the
complete discussion of myelodysplastic syndrome. 2000 U.S. standard population census.
592 PART VI Hematology in Selected Populations

fibrinolytic factors. These changes are thought to contribute to association between factor VII and venous thrombosis have
the increased incidence of thrombosis in the elderly. The rate yielded conflicting findings.173
of venous thromboembolism, for example, increases from 1 PAI-1, the major inhibitor of fibrinolysis, increases with
per 10,000 in the young (under 40 years of age) to 1 per 1000 aging. PAI-1 has been shown to promote age-dependent
in the elderly (older than 75 years).172 thrombosis in animal models and could play an important role
Fibrinogen, factor V, factor VII, factor VIII, factor IX, factor in causing hypercoagulability in the elderly.176-178
XIII, high-molecular-weight kininogen, and prekallikrein Platelets increase in activity with age, as evidenced by a
increase in healthy individuals as they age.173 Fibrinogen, decrease in bleeding time as males age from 11 to 70 years179
which has been implicated as a primary risk factor for throm- and an increase in markers of platelet activation, platelet -
botic disorders, increases approximately 10mg/dL per decade thromboglobulin and platelet factor 4.180 Increased platelet
in the elderly (65 to 79 years),174 from 280mg/dL to over activity with aging is also associated with increased platelet
300mg/dL. Elevated levels of factor VIII also have been associ- phospholipids, which suggests an age-related increase in
ated with increased risk of venous thrombosis.175 Studies of the platelet transmembrane signalling.181

SUMMARY
The newborn infant, preadolescent child, and elderly adult exhibit There is a gradual decline in hemoglobin starting at middle age,
profound hematologic differences from one another. and the mean level decreases by about 1g/dL during the sixth
Hematologic parameters continue to change and evolve over the through eighth decades.
first few days, weeks, and months of life. Data must be assessed Although anemia is common in elderly patients, it is not a normal
in light of gestational age, birth weight, and developmental differ- occurrence in the aging process. The cause of anemia may be
ences between newborns and older infants. multifactorial in elderly patients.
The erythrocytes of newborn infants are markedly macrocytic at Anemia of chronic inflammation and iron deficiency anemia are
birth. A condition known as physiologic anemia of infancy occurs the most common anemias seen in the elderly.
after the first weeks of life. Infants born prematurely also experi- Vitamin B12 deficiency and folate deficiency are other causes of
ence a decrease in hemoglobin concentration, which is termed anemia in the elderly.
physiologic anemia of prematurity. Immunosenescence refers to the adverse changes in the immune
Iron deficiency is the most frequent cause of anemia in children. system associated with aging.
Fluctuations in the number of WBCs are common at all ages but The elderly experience an increased frequency of many neoplastic
are greatest in infants. Leukocytosis is typical at birth for full-term and malignant disorders, such as acute and chronic leukemia,
and preterm infants, with a mean of 22 109 cells/L (range, 9 to myelodysplastic syndromes, and myeloproliferative neoplasms.
30 109 cells/L) at 12 hours of life. There is an excess of seg- The elderly are at an increased risk of thrombosis associated
mented neutrophils, bands, and occasional metamyelocytes with with age-related changes in the vascular and hemostatic
no evidence of disease. systems.
Sepsis in neonates is a common cause of morbidity, particularly
in premature and low-birth-weight infants. Defective B-cell Now that you have completed this chapter, go back and
response against polysaccharide agents as well as abnormal cyto- read again the case study at the beginning and respond
kine release by neutrophils and monocytes has been implicated. to the questions presented.
Although hemostatic values are different in infants and children
from those in adults, this population is not at increased risk of
bleeding or thrombosis.

R E V I E W Q UESTIONS
1. The CBC results for children (age 3 to 12 years) differ from 2. Physiologic anemia of infancy results from:
those of adults chiefly in what respect? a. Iron deficiency caused by a milk-only diet during the
a. NRBCs are present. early neonatal period
b. Notable polychromasia is seen, indicating increased b. Increased oxygenation of blood and decreased
reticulocytosis. erythropoietin
c. Platelet count is lower. c. Replacement of active marrow with fat soon after
d. The percentage of lymphocytes is higher. birth
d. Hb F and its diminished oxygen delivery to tissues
CHAPTER 38 Pediatric and Geriatric Hematology 593

3. The CBC report on a 3-day-old neonate who was born 6 7. Which of the following are the most common anemias in
weeks prematurely shows a decrease in hemoglobin com- the elderly population?
pared with the value obtained 2 days earlier. Which of the a. Megaloblastic anemia and iron deficiency anemia
following should be considered as an explanation for this b. Sideroblastic anemia and megaloblastic anemia
result when no apparent source of hemolysis or bleeding c. Myelophthisic anemia and anemia of chronic
is evident? inflammation
a. The sample was collected from a vein at the time that d. Iron deficiency anemia and anemia of chronic
an intravenous line was inserted. inflammation
b. The sample was collected by heel puncture rather
than finger puncture because of the infants small 8. When iron deficiency is recognized in an elderly individ-
size. ual, the cause is usually:
c. The umbilical cord was clamped quickly to begin a. An iron-deficient diet
appropriate treatment for a preterm infant. b. Gastrointestinal bleeding
d. The infant has become dehydrated. c. Diminished absorption
d. Impaired incorporation of iron into heme as a result
4. Morphologically, the hematogones in newborns are: of telomere loss
a. Similar to those seen in megaloblastic anemia
b. Easily confused with leukemic blasts 9. Which of the following conditions is least likely in an
c. Monocytoid in appearance elderly individual?
d. Similar to adult lymphocytes a. Acute lymphoblastic leukemia
b. Multiple myeloma
5. The most frequent cause of anemia in childhood is: c. Myelodysplasia
a. Vitamin B12 deficiency d. Chronic lymphocytic leukemia
b. Drug-related hemolysis
c. Iron deficiency 10. The multiple medications used by the elderly makes this
d. Folate deficiency population more prone to:
a. Anemia of chronic disease
6. As age increases, the hemoglobin level of elderly adults: b. Megaloblastic anemia
a. Remains unchanged from that of middle-aged c. Hemolytic anemia
adults d. Iron deficiency anemia
b. Increases due to diminished respiration and poor
tissue oxygenation
c. Decreases for reasons that are unclear
d. Becomes comparable to that of newborns

REFERENCES
1. Brugnara C, Platt OS: The neonatal erythrocyte and its dis- 8. Christensen RD: Circulating pluripotent hematopoietic pro-
orders. In Orkin SH, Nathan DG, Ginsburg D, et al, editors: genitor cells in neonates. J Pediatr 110:622-625, 1987.
Nathan and Oskis hematology of infancy and childhood, ed 7, 9. Yoder MC: Embryonic hematopoiesis. In Christensen RD,
Philadelphia, 2009, Saunders, pp 21-66. editor: Hematologic problems of the neonate, Philadelphia,
2. Segal GB, Palis J: Hematology of the newborn. In Lichtman 2002, Saunders, pp 3-20.
MA, Beutler E, Kipps TJ, et al, editors: Williams hematology, 10. Tavassoli M: Embryonic and fetal hemopoiesis: an over-
ed 7, New York, 2006, McGraw-Hill, pp 81-98. view. Blood Cells 1:269-281, 1991.
3. Hann IM, Bodger MP, Hoffbrand AV: Development of plu- 11. Adams-Chapman I: The high risk infant. In Kliegman
ripotent hematopoietic progenitor cells in the human fetus. RM, Behrman RE, Jenson HB, et al, editors: Nelson
Blood 62:118-123, 1983. textbook of pediatrics, ed 18, Philadelphia, 2007, Saunders,
4. Forestier F, Daffos F, Catherine N, et al: Developmental pp 698-711.
hematopoiesis in normal human fetal blood. Blood 12. Peterec SM, Warshaw JB: The premature newborn. In
1991;77:2360-2363. McMillan JA, editor: Oskis pediatrics, ed 4, Philadelphia,
5. Ohls RK, Christensen RD: Development of the hematopoi- 2006, Lippincott Williams & Wilkins, pp 220-235.
etic system. In Behrman RE, Kliegman R, Jenson HB, editors: 13. Miller DR: Normal blood values from birth through adoles-
Nelson textbook of pediatrics, ed 17, Philadelphia, 2004, cence. In Miller DR, Baehner RL, editors: Blood diseases of
Saunders, pp 1599-1602. infancy and childhood: in the tradition of CH Smith, ed 7,
6. Christensen RD: Hematopoiesis in the fetus and neonate. St Louis, 1995, Mosby, pp 3-53.
Pediatr Res 26:531-535, 1989. 14. Oski FA, Naiman JL: Normal blood values in the newborn
7. Linch DC, Knott LJ, Rodeck CH, et al: Studies of circulating period. In Oski FA, Naiman JL, editors: Hematologic problems
hematopoietic progenitor cells in human fetal blood. Blood in the newborn, ed 3, Philadelphia, 1982, WB Saunders,
59:976-979, 1982. pp 1-31.
594 PART VI Hematology in Selected Populations

15. Speakman ED, Boyd JC, Bruns DE: Measurement of methe- 36. McIntosh N, Kempson C, Tyler RM: Blood counts in
moglobin in neonatal samples containing fetal hemoglo- extremely low birth weight infants. Arch Dis Child 63:74-76,
bin. Clin Chem 41:458-461, 1995. 1988.
16. Puukka R, Puukka M: Effect of hemoglobin F on measure- 37. Cavaliere TA: Red blood cell indices: implications for prac-
ments of hemoglobin A1c with physicians office analyzers. tice. Newborn Infant Nurs Rev 4:231-239, 2004.
Clin Chem 40:342-343, 1994. 38. Obladen M, Diepold K, Maier RF: Venous and arterial
17. Sandberg S, Sonstabo K, Christensen G: Influence of lipid hematologic profiles of very low birth weight infants. Pedi-
and leukocytes on the haemoglobin determination by atrics 106:707-711, 2000.
Coulter Counter S Plus III, Technicon H 6000, Technicon 39. Halvorsen S, Finne PH: Erythropoietin reduction in the
H1, LK 540, Reflotron and Hemocap. Scand J Clin Lab Invest human fetus and newborn. Ann N Y Acad Sci 149:576-577,
49:145-148, 1989. 1968.
18. Geaghan SM: Hematologic values and appearances in the 40. Mann DL, Sites ML, Donati RM: Erythropoietic stimulating
healthy fetus, neonate, and child. Clin Lab Med 19:1-37, activity during the first ninety days of life. Proc Soc Exp Biol
1999. Med 118:212-214, 1965.
19. Matoth Y, Zaizov R, Varsano I: Postnatal changes in some 41. Newborn Services Clinical Guideline: Normal haematologi-
red cell parameters. Acta Paediatr Scand 60:317-323, 1971. cal values. Available at: http://www.adhb.govt.nz/newborn/
20. Christensen RD: Expected hematologic values for term and Guidelines/blood/haematologicalvalues.htm. Accessed July
preterm neonates. In Christensen RD, editor: Hematologic 10, 2010.
problems of the neonate, Philadelphia, 2000, Saunders, 42. Jopling J, Henry E, Wiedmeier SE, et al: Reference ranges for
pp 117-136. hematocrit and blood hemoglobin concentration during
21. Zaizov R, Matoth Y: Red cell values on the first postnatal the neonatal period: data from a multihospital health care
day during the last 16 weeks of gestation. Am J Hematol system. Pediatrics 123:e333-e337, 2009.
1:275-278, 1976. 43. Means RT, Glader B: Anemia: general considerations.
22. Thomas RM, Canning CE, Cotes PM, et al: Erythropoietin In Greer JP, Foerster J, Rodgers GM, et al, editors:
and cord blood hemoglobin in the regulation of human Wintrobes clinical hematology, ed 12, Philadelphia, 2009,
fetal erythropoiesis. Br J Obstet Gynaecol 90:795-800, 1983. Wolters Kluwer Health/Lippincott Williams & Wilkins,
23. Pahal GS, Jauniaux E, Kinnon C, et al: Normal development pp 779-809.
of human fetal hematopoiesis between eight and seventeen 44. Nickerson HJ, Silberman T, Park RW, et al: Treatment
weeks gestation. Am J Obstet Gynecol 183:1029-1034, of iron deficiency anemia and associated protein-losing
2000. enteropathy in children. J Pediatr Hematol Oncol 22:50-54,
24. Luchtman-Jones LL, Schwartz AL, Wilson DB: Hematologic 2000.
problems in the fetus and neonate. In Martin RJ, Fanaroff 45. Dagnelie PC, Van Staveren WA, Hautvast JG: Stunting and
AA, Walsh MC, editors: Neonatal-perinatal medicine, ed 8, nutrient deficiencies in children on alternative diets. Acta
St Louis, 2006, Mosby, pp 1287-1344. Paediatr Scand 374:111-118, 1991.
25. Soldin S, Brugnara C, Wong EC, editors: Pediatric reference 46. Schneede J, Dagnelie PC, Van Staveren WA, et al: Methyl-
intervals, ed 6, Washington, DC, 2007, AACC Press. malonic acid and homocysteine in plasma as indicators of
26. Luchtman-Jones L, Schwartz AL, Wilson DB: The blood and functional cobalamin deficiency in infants on macrobiotic
hematopoietic system: hematologic problems of the fetus diets. Pediatr Res 36:194-201, 1994.
and neonate. In Martin RJ, Fanaroff AA, Walsh MC, editors: 47. Walter T, DeAndraca I, Chadud P, et al: Iron deficiency
Neonatal-perinatal medicine, ed 8, Philadelphia, 2006, anemia: adverse effects on infant psychomotor develop-
Mosby, pp 1287-1344. ment. Pediatrics 84:7-17, 1989.
27. Zipursky MD, Brown E, Palko J, et al: The erythrocyte dif- 48. Bridges KR, Pearson HA: Anemias and other red cell disorders.
ferential count in newborn infants. Am J Pediatr Hematol New York, 2008, McGraw-Hill, pp 99-100.
Oncol 3:45-51, 1983. 49. Will AM: Disorders of iron metabolism: iron deficiency, iron
28. Cook CD, Brodie HR, Allen DW: Measurement of fetal overload and the sideroblastic anemias. In Arceci RJ, Hann
hemoglobin in premature infants: correlation with gesta- IM, Smith OP, editors: Pediatric hematology, ed 3, Malden,
tional age and intrauterine hypoxia. Pediatrics 20:272-278, Mass, 2006, Blackwell, pp 79-104.
1957. 50. Andrews NC, Ulrich CK, Fleming MD: Disorders of iron
29. Bard H: The postnatal decline of hemoglobin F in normal metabolism and sideroblastic anemia. In Orkin SH, Nathan
full-term infants. J Clin Invest 55:395-398, 1975. DG, Ginsburg D, et al, editors: Nathan and Oskis hematology
30. Bard H: Postnatal fetal and adult hemoglobin synthesis in of infancy and childhood, ed 7, Philadelphia, 2009, Saunders,
early preterm newborn infants. J Clin Invest 52:1789-1795, pp 521-570.
1973. 51. Looker A: Iron deficiencyUnited States, 1999-2000.
31. Dallman PR, George DB, Allen CM, et al: Hemoglobin con- MMWR Morb Mortal Wkly Rep 51:897-899, 2002.
centration in white, black, and oriental children: is there a 52. Gartner LM, Morton J, Lawrence RA, et al; American
need for separate criteria in screening for anemia? Am J Clin Academy of Pediatrics Section on Breastfeeding: Breastfeed-
Nutr 31:377-380, 1978. ing and the use of human milk. Pediatrics 115:496-506,
32. Doyle JJ: The role of erythropoietin in the anemia of pre- 2005.
maturity. Semin Perinatol 21:20-27, 1997. 53. Kanakoudi F, Drossou V, Tzimouli V, et al: Serum concentra-
33. Strauss RG: Recombinant erythropoietin for the anemia of tions of 10 acute-phase proteins in health term and preterm
prematurity: still a promise, not a panacea. J Pediatr 131:653- infants from birth to age 6 months. Clin Chem 41:605-608,
655, 1997. 1995.
34. Garby L, Sjlin S, Vuille J: Studies of erythrokinetics in 54. Minder N, Cohn J: Serum iron, serum transferrin and trans-
infancy. III. Disappearance from plasma and red cell uptake ferrin saturation in healthy children without iron deficiency.
of radioactive iron injected intravenously. Acta Paediatr Eur J Pediatr 143:96-98, 1984.
52:537-553, 1963. 55. Saarinen UM, Siimes MA: Serum ferritin in assessment of
35. OBrien RT, Pearson HA: Physiologic anemia of the newborn iron nutrition in healthy infants. Acta Paediatr Scand 67:745-
infant. J Pediatr 79:132-138, 1971. 751, 1978.
CHAPTER 38 Pediatric and Geriatric Hematology 595
56. Siimes MA, Addiego JE, Dall PR: Ferritin in serum: diagnosis 75. Squire E, Favara B, Todd J: Diagnosis of neonatal bacterial
of iron deficiency and iron overload in infants and children. infection: hematologic and pathologic findings in fatal and
Blood 43:581-590, 1974. nonfatal cases. Pediatrics 64:60-64, 1979.
57. Cairo MS, Bracho F: White blood cells. In Rudolph CD, 76. Garra G, Cunningham SJ, Crain EF: Reappraisal of criteria
Rudolph AM, Hostetter MK, et al, editors: Rudolphs pediat- used to predict serious bacterial illness in febrile infants less
rics, ed 21, New York, 2003, McGraw-Hill, pp 1547-1558. than 8 weeks of age. Acad Emerg Med 12:921-925, 2005.
58. Schmutz N, Henry E, Jopling J, et al: Expected ranges for 77. Jaskiewicz JA, McCarthy CA, Richardson AC, et al: Febrile
blood neutrophil concentrations of neonates: the Manroe infants at low risk for serious bacterial infectionan
and Mouzinho charts revisited. J Perinatol 28:275-281, appraisal of the Rochester criteria and implications for man-
2008. agement. Febrile Infant Collaborative Study Group. Pediat-
59. Christensen RD, Henry E, Jopling J, et al: The CBC: reference rics 94:390-396, 1994.
ranges for neonates. Semin Perinatol 33:3-11, 2009. 78. Timens W, Rozeboom T, Poppema S: Fetal and neonatal
60. Caramihai E, Karayalein G, Aballi AJ, et al: Leukocyte count development of human spleen: an immunohistological
differences in health white and black children 1 to 5 years study. Immunology 60:603-609, 1987.
of age. J Pediatr 86:252-254, 1975. 79. Davidson D, Miskolci V, Clark DC, et al: Interleukin-10
61. Mouzinho A, Rosenfeld CR, Sanchez PJ, et al: Revised refer- production after pro-inflammatory stimulation of neutro-
ence ranges for circulating neutrophils in very-low-birth- phils and monocytic cells of the newborn. Neonatology
weight neonates. Pediatrics 94:76-82, 1994. 92:127-133, 2007.
62. Alexander GR, Kogan M, Bader G, et al: US birth weight/ 80. Rondini G, Chirico G: Hematopoietic growth factor levels
gestational agespecific neonatal mortality: 1995-1997 rates in term and preterm infants. Curr Opin Hematol 6:192-197,
for whites, Hispanics and blacks. Pediatrics 111:e61-e66, 1999.
2003. 81. Kotiranta-Ainamo A, Rautonen J, Rautonen N: Imbalanced
63. Monroe BL, Weinberg AG, Rosenfeld CR: The neonatal cytokine secretion in newborns. Biol Neonate 85;55-60,
blood count in health and disease: I. reference values for 2004.
neutrophilic cells. J Pediatr 95:89-98, 1976. 82. Akenzua GI, Hui YT, Milner R, et al: Neutrophil and band
64. Dinauer MC, Newburger PE: The phagocyte system and counts in the diagnosis of neonatal infection. Pediatrics
disorders of granulopoiesis and granulocyte function. In 54:38-42, 1974.
Orkin SH, Nathan DG, Ginsburg D, et al, editors: Nathan 83. Bhandari V, Wang C, Rinder C, et al: Hematologic profile of
and Oskis hematology of infancy and childhood, ed 7, sepsis in neonates: neutrophil CD64 as a diagnostic marker.
Philadelphia, 2009, Saunders, pp 1109-1217. Pediatrics 121:129-134, 2008.
65. Ezekowitz RAB: Hematologic manifestations of systemic 84. Rey C, Los Arcos M, Concha A, et al: Procalcitonin and
diseases. In Orkin SH, Nathan DG, Ginsburg D, et al, C-reactive protein as markers of systemic inflammatory
editors: Nathan and Oskis hematology of infancy and child- response syndrome severity in critically ill children. Intensive
hood, ed 7, Philadelphia, 2009, Saunders, pp 1680-1739. Care Med 33:477-484, 2007.
66. Wright B, McKenna RW, Asplund SL, et al: Maturing B-cell 85. Fogel BJ, Arais D, Kung F: Platelet counts in healthy prema-
precursors in bone marrow: a detailed subset analysis of ture infants. J Pediatr 73:108-110, 1968.
141 cases by 4-color flow cytometry. Mod Pathol 15:270A, 86. Sell EJ, Corrigan JJ: Platelet counts, fibrinogen concentra-
2002. tions and factor V and factor VIII levels in healthy infants
67. Rimsza LM, Douglas VK, Tighe P, et al: Benign B-cell precur- according to gestational age. J Pediatr 82:1028-1032, 1973.
sors (hematogones) are the predominant lymphoid popula- 87. Mehta P, Vasa R, Neumann L, et al: Thrombocytopenia in
tion in the bone marrow of preterm infants. Biol Neonate the high-risk infant. J Pediatr 97:791-794, 1980.
86:247-253, 2004. 88. Meberg A, Halvorsen S, Orstavik I: Transitory thrombocyto-
68. Vogel P, Erf LA: Hematological observations on bone penia in small-for-dates infants, possibly related to mater-
marrow obtained by sternal punctures. Am J Clin Pathol nal smoking. Lancet 2:303-304, 1977.
7;436-447, 1937. 89. Andrew M, Paes B, Milner R, et al: Development of the
69. Muehleck SD, McKenna RW, Gale PF, et al: Terminal deoxy- human coagulation system in the full-term infant. Blood
nucleotidyl transferase (TdT)positive cells in bone marrow 70:165-172, 1987.
in the absence of hematologic malignancy. Am J Clin Pathol 90. Andrew M, Vegh P, Johnston M, et al: Maturation of the
79:277-284, 1983. hemostatic system during childhood. Blood 80:1998-2005,
70. Brunning RD, McKenna RW: Tumors of the bone 1992.
marrow, Washington DC, 1994, Armed Forces Institute of 91. Manno CS: Management of bleeding disorders in children.
Pathology. Hematology Am Soc Hematol Educ Program 416-422, 2005.
71. McKenna RW, Washington LT, Aquino DB, et al: Immuno- 92. Pabinger I, Schneider B: Thrombotic risk in hereditary anti-
phenotypic analysis of hematogones (B-lymphocyte precur- thrombin III, protein C, or protein S deficiency. A coopera-
sors) in 662 consecutive bone marrow specimens by 4-color tive, retrospective study. Gesellschaft fr Thrombose- und
flow cytometry. Blood 98:2498-2507, 2001. Hamostaseforschung (GTH) Study Group on Natural Inhib-
72. Intermesoli T, Mangili G, Salvi A, et al: Abnormally itors. Arterioscler Thromb Vasc Biol 12:742-748, 1996.
expanded pro-B hematogones associated with congenital 93. Worly FM, Fortenberry JD, Hansen I, et al: Deep venous
cytomegalovirus infection. Am J Hematol 82:934-936, 2007. thrombosis in children with diabetic ketoacidosis and
73. Chirico G, Ciardelli L, Gasparoni A: Clinical and experimen- femoral central venous catheters. Pediatrics 113:57-60, 2004.
tal aspects on the use of granulocyte colony stimulating 94. Gutierrez JA, Bagatell R, Samson MP, et al: Femoral central
factor (G-CSF) in the neonate. In Bellanti JA, Bracci R, venous catheterassociated deep venous thrombosis in chil-
Prindull G, et al, editors: Neonatal hematology and immuno dren with diabetic ketoacidosis. Crit Care Med 31:80-83,
logy III, Amsterdam, 1997, Elsevier Science, pp 33-38. 2003.
74. Weimann E, Rutkowski S, Reisbach G: G-CSF, GM-CSF and 95. Ruud E, Holmstrom II, Natvig S, et al: Prevalence of throm-
IL-6 levels in cord blood: diminished increase of G-CSF and bophilia and central venous catheterassociated neck vein
IL-6 in preterms with perinatal infection compared to term thrombosis in 41 children with cancer: a prospective study.
neonates. J Perinat Med 26:211-218, 1998. Med Pediatr Oncol 38:405-410, 2002.
596 PART VI Hematology in Selected Populations

96. Parasuraman S, Goldhaber SZ: Venous thromboembolism 119. Song L, Kim YH, Chopra RK, et al: Age related effects in T
in children. Circulation 113:e12-e16, 2006. cell activation and proliferation. Exp Gerontol 28:313-321,
97. Federal Interagency Forum on Aging Related Statistics: 1993.
Older Americans 2008: key indicators of well-being, 2008. 120. Wick G, Grubeck-Lobenstein B: The aging immune system:
Available at: http://www.AgingStats.gov. Accessed July 10, primary and secondary alterations of immune reactivity in
2010. the elderly. Exp Gerontol 32:401-413, 1997.
98. US Department of Commerce, Economics and Statistics 121. Kawai T, Akira S: TLR signaling. Semin Immunol 19:24-32,
Administration, Bureau of the Census: We the American 2007.
elderly, 1993. Available at: http://www.census.gov/apsd/ 122. Sansoni P, Cossarizza A, Brianti V, et al: Lymphocyte subsets
wepeople/we-9.pdf. Accessed July 10, 2010. and natural killer cell activity in healthy old people and
99. Zauber NP, Zauber AG: Hematologic data of healthy old centenarians. Blood 82:2767-2773, 1993.
people. JAMA 257:2181-2184, 1987. 123. Grubeck-Lobenstein B: Changes in the aging immune
100. Chatta GS, Dale DC: Aging and haemopoiesis: implications system. Biologicals 25:205-208, 1997.
for treatment with haemopoietic growth factors. Drugs 124. Tefferi A, Thiele J, Orazi A, et al: Proposals and rationale for
Aging 9:37-47, 1996. revision of the World Health Organization diagnostic crite-
101. Lansdorp PM: Self-renewal of stem cells. Biol Blood Marrow ria for polycythemia vera, essential thrombocythemia, and
Transplant 3:171-178, 1997. primary myelofibrosis: recommendation from an ad hoc
102. Hartstock RJ, Smith EB, Petty CS: Normal variation with international expert panel. Blood 110:1092-1097, 2007.
aging of the amount of hematopoietic tissue in bone 125. Zahavi J, Jones NA, Leyton J, et al: Enhanced in vivo platelet
marrow from the anterior iliac crest. Am J Clin Pathol release reaction in old healthy individuals. Thromb Res
43:326-331, 1965. 17:329-336, 1980.
103. Ricci C, Cova M, Kang Y, et al: Normal age-related pattern 126. Chong BH: Heparin-induced thrombocytopenia. Br J Hae-
of cellular and fatty bone marrow distribution in the axial matol 89:431-439, 1995.
skeleton: MR imaging study. Radiology 177:83-88, 1990. 127. Cortelazzo S, Finazzi G, Ruggeri M, et al: Hydroxyurea for
104. Frenck RW, Jr, Blackburn EH, Shannon KM: The rate of patients with essential thrombocythemia and a high risk for
telomere sequence loss in human leukocyte varies with age. thrombosis. N Engl J Med 332:1132-1136, 1995.
Proc Natl Acad Sci U S A 95:5607-5610, 1998. 128. Balducci L: Anemia, cancer, and aging. Cancer Control
105. Geiger H, Rudolph KL: Aging in the lympho-hematopoietic 10:478-486, 2003.
stem cell compartment. Trends Immunol 30:360-365, 2009. 129. Smith D: Anemia in the elderly. Am Fam Physician 62:1565-
106. Nilsson-Ehle H, Jagenburg R, Landahl S, et al: Blood haemo- 1571, 2000.
globin values in the elderly: implications for reference inter- 130. Izaka GJ, Westendorp RG, Knook DL: The definition of
vals from age 70 to 88. Eur J Haematol 65:296-305, 2000. anemia in older persons. JAMA 281:1714-1717, 1999.
107. Myers AM, Saunders CR, Chalmers DG: The haemoglobin 131. Lipschitz D: Medical and functional consequences of
level of fit elderly people. Lancet 2:261-263, 1968. anemia in the elderly. J Am Geriatr Soc 51(suppl):10-13,
108. Salive ME, Cornoni-Huntley J, Guralnik JM, et al: Anemia 2003.
and hemoglobin level in older persons: relationship with 132. Guralnik J, Eisenstaedt R, Ferrucci L, et al: Prevalence of
age, gender and health status. J Am Geriatr Soc 40:489-496, anemia in persons 65 years and older in the United States:
1992. evidence for a high rate of unexplained anemia. Blood
109. Allan RN, Alexander MK: A sex difference in the leukocyte 104:2263-2268, 2004.
count. J Clin Pathol 21:691-694, 1965. 133. Begh C, Wilson A, Ershler WB: Prevalence and outcomes
110. Shayne M, Lichtman MA: Hematology in older persons. of anemia in geriatrics: a systematic review of the literature.
In Lichtman MA, Beutler E, Kipps TJ, et al, editors: Am J Med 116:3S-10S, 2004.
Williams hematology, ed 7, New York, 2006, McGraw-Hill, 134. Howe RB: Anemia in the elderly. Postgrad Med 73:153-160,
pp 111-121. 1983.
111. Panda A, Arjona A, Sapey E, et al: Human innate immunose- 135. Timiras ML, Brownstein H: Prevalence of anemia and cor-
nescence: causes and consequences for immunity in old age. relation of hemoglobin with age in a geriatric screening
Trends Immunol 30:325-333, 2009. clinic population. J Am Geriatr Soc 35:639-643, 1987.
112. Fulop T, Pawalec G, Castle S, Loeb M: Immunosenescence 136. Ania BJ, Suman VJ, Fairbanks VF, et al: Prevalence of anemia
and vaccination in nursing home residents. Clin Infect Dis in medical practice: community versus referral patients.
48:443-448, 2009. Mayo Clin Proc 69:730-735, 1994.
113. Fulop T, Larbi A, Wikby A, et al: Dysregulation of T cell 137. Smith D: Management and treatment of anemia in the
function in the elderly: scientific basis and clinical implica- elderly. Clin Geriatr 10:8, 2002.
tions. Drugs Aging 22:589-603, 2005. 138. Daly MP: Anemia in the elderly. Am Fam Physician 39:129-
114. Mangolas SC, Jilka RL: Bone marrow, cytokines and bone 136, 1989.
remodeling. N Engl J Med 332:302-311, 1995. 139. Walsh JR: Hematologic problems. In Cassel CK, Cohen HJ,
115. Globerson A: T lymphocytes and aging. Int Arch Allergy Larson EB, et al, editors: Geriatric medicine, ed 3, New York,
Immunol 107:491-497, 1995. 1997, Springer Verlag, pp 627-636.
116. Nel AE, Slaughter N: T-cell activation through the antigen 140. Lipschitz DA: The anemia of chronic disease. J Am Geriatr
receptor, part 2: role of signaling cascades in T-cell differen- Soc 38:1258-1264, 1990.
tiation, anergy, immune senescence, and development of 141. Gardner LB, Benz EJ: Anemia of chronic diseases. In Hoffman
immunotherapy. J Allergy Clin Immunol 109:901-915, 2002. R, Furie B, Benz EJ, Jr, et al, editors: Hematology basic principles
117. Larbi A, Douziech N, Dupuis G, et al: Age-associated altera- and practices, ed 5, Philadelphia, 2009, Saunders.
tions in the recruitment of signal transduction proteins to 142. Cash J, Sears DA: The anemia of chronic disease: spectrum
lipid rafts in human T lymphocytes. J Leukoc Biol 75:373- of associated disease in a series of unselected hospitalized
381, 2004. patients. Am J Med 87:638-644, 1989.
118. Larbi A, Dupuis G, Khalil A, et al: Differential role of lipid 143. Joosten E: Strategies for the laboratory diagnosis of some
rafts in the functions of CD4+ and CD8+ human T lympho- common causes of anaemia in elderly patients. Gerontology
cytes with aging. Cell Signal 18:1017-1030, 2006. 50:49-56, 2004.
CHAPTER 38 Pediatric and Geriatric Hematology 597
144. Ania BJ, Suman VJ, Fairbanks VF, et al: Prevalence of anemia 164. Pfeiffer CM, Johnson CL, Jain RB, et al: Trends in blood
in medical practice: community versus referral patients. folate and vitamin B-12 concentrations in the United States,
Mayo Clin Proc 69:730-735, 1994. 1988-2004. Am J Clin Nutr 86:718-727, 2007.
145. Means RT: The anaemia of infection. Baillieres Clin Haematol 165. Andrews NC, Schmidt PJ: Iron homeostasis. Annu Rev
13:151-162, 2000. Physiol 69:69-85, 2007.
146. Smith DI: Anemia in the elderly. Am Fam Physician 62:1565- 166. Lindenbaum J: Folate and vitamin B12 deficiencies in alco-
1572, 2000. holism. Semin Hematol 17:119-128, 1980.
147. Chiari MM, Bagnoli R, DeLuca P, et al: Influence of acute 167. Petz LD: Drug-induced immune haemolytic anaemia. Clin
inflammation on iron and nutritional status indexes in Haematol 9:455-482, 1980.
older inpatients. J Am Geriatr Soc 43:767-771, 1995. 168. Saba HA: Myelodysplastic syndromes in the elderly.
148. Daly MP: Anemia in the elderly. Am Fam Physician 39:129- Cancer Control 8(1):79-102, 2001. Available at: http://
136, 1989. www.medscape.com/viewarticle/409029. Accessed July 10,
149. Pennypacker LC, Allen RH, Kelly JP, et al: High prevalence 2010.
of cobalamin deficiency in elderly outpatients. J Am Geriatr 169. Altekruse SF, Kosary CL, Krapcho M, et al, editors: SEER
Soc 40:1197-1204, 1992. cancer statistics review, 1975-2007, Bethesda, Md, 2010,
150. Stabler SP: Vitamin B12 deficiency in older people: improv- National Cancer Institute. Based on November 2009 SEER
ing diagnosis and preventing disability. J Am Geriatr Soc data submission, posted to the SEER website 2010. Avail-
46:1317-1319, 1998. able at: http://seer.cancer.gov/csr/1975_2007/. Accessed
151. Nexo E, Hansen M, Rasmussen K, et al: How to diagnosis July 10, 2010.
cobalamin deficiency. Scand J Clin Lab Invest Suppl 219:61- 170. Leukemia Lymphoma Society: Disease information. Avail-
76, 1994. able at: http://www.leukemia-lymphoma.org. Accessed July
152. Powell DE, Thomas JH: The iron binding capacity of serum 10, 2010.
in elderly hospital patients. Gerontol Clin (Basel) 11:36-47, 171. American Cancer Society website. Available at: http://
1969. www.cancer.org/docroot/home/index.asp?level=0. Accessed
153. Healton EB, Savage DG, Brust JC, et al: Neurologic aspects July 10, 2010.
of cobalamin deficiency. Medicine 70:229-244, 1991. 172. Martinelli I: Risk factors for venous thromboembolism.
154. Savage DG, Lindenbaum J: Neurological complications of Thromb Haemost 86:395-403, 2001.
acquired cobalamin deficiency: clinical aspects. Baillieres 173. Franchini M: Hemostasis and aging. Crit Rev Oncol Hematol
Clin Haematol 8:657-678, 1995. 60:144-151, 2006.
155. Matthews JH: Cobalamin and folate deficiency in the 174. Kannel WB, Wolf PA, Castelli WP, et al: Fibrinogen and risk
elderly. Baillieres Clin Haematol 54:245-253, 1999. of cardiovascular disease. The Framingham Study. JAMA
156. Doscherholmen A, Swaim WR: Impaired assimilation of egg 258:1183-1186, 1987.
57
Co vitamin B12 in patients with hypochlorhydria and 175. Kster T, Blann D, Brit E, et al: Role of clotting factor VIII
achlorhydria and after gastric resection. Gastroenterology and effect of von Willebrand factor on occurrence of deep-
64:913-919, 1973. vein thrombosis. Lancet 345:152-155, 1995.
157. Suter PM, Golner BB, Goldin BR, et al: Reversal of protein- 176. Cushman M, Lemaitre RN, Kuller LH, et al: Fibrinolytic
bound vitamin B12 malabsorption with antibiotics in atro- activation markers predict myocardial infarction in the
phic gastritis. Gastroenterology 101:1039-1045, 1991. elderly. The Cardiovascular Health Study. Arterioscler Thromb
158. Baik HW, Russell RM: Vitamin B12 deficiency in the elderly. Vasc Biol 19:493-498, 1999.
Annu Rev Nutr 19:357-377, 1999. 177. Yamamoto K, Takeshita K, Shimokawa T, et al: Plasminogen
159. Carmel R: Prevalence of undiagnosed pernicious anemia in activator inhibitor-1 is a major stress-regulated gene: impli-
the elderly. Arch Intern Med 156:1097-1100, 1996. cations for stress-induced thrombosis in aged individuals.
160. Norman EJ, Morrison JA: Screening elderly populations for Proc Natl Acad Sci U S A 99:890-895, 2002.
cobalamin (vitamin B12) deficiency using the urinary methy- 178. Yamamoto K, Takeshita K, Kojima T, et al: Aging and plas-
malonic acid assay by gas chromatography mass spectrom- minogen activator inhibitor-1 (PAI-1) regulation: implica-
etry. Am J Med 94:489-494, 1993. tion in the pathogenesis of thrombotic disorders in the
161. Loikas S, Koskinen P, Irjala K, et al: Vitamin B12 deficiency elderly. Cardiovasc Res 66:276-285, 2005.
in the aged: a population-based study. Age Ageing 36:177- 179. Jrgensen KA, Dyerberg J, Olesen AS, et al: Acetylsalicylic
183, 2007. acid, bleeding time and age. Thromb Res 19:799-805,
162. Nierodzik ALR, Sutin D, Freedman ML: Blood disorders and 1980.
their management in old age. In Tallis RC, Fillit HM, editors: 180. Zahavi J, Jones NA, Leyton J, et al: Enhanced in vivo platelet
Brocklehursts textbook of geriatric medicine and gerontology, release reaction in old healthy individuals. Thromb Res
ed 6, London, 2003, Elsevier Science, pp 1229-1268. 17:329-336, 1980.
163. Karnaze DS, Carmel R: Low serum cobalamin levels in 181. Bastyr EJ, 3rd, Kadrofske MM, Vinik AI: Platelet activity and
primary degenerative dementia. Arch Intern Med 17:429- phosphoinositide turnover increase with advancing age. Am
431, 1987. J Med 88:601-606, 1990.
PART VII Cell-Counting Automation

39 Automated Cell-Counting
Instrumentation
Sharral Longanbach, Deanne H. Chapman,* and Martha K. Miers

OUTLINE OBJECTIVES
General Principles of After completion of this chapter, the reader will be able to:
Hematology
Instrumentation 1. Explain the different principles of automated cell 6. Interpret and compare patient data, including
Electronic Impedance counting. WBC and red blood cell histograms or cytograms
Radiofrequency 2. Describe how the general principles are implemented or both, obtained from the major hematology
Optical Scatter on the different instruments discussed. instruments.
Principal Cell-Counting 3. Identify the hemogram parameters directly measured 7. Explain the general principles of automated reticulo-
Instruments on the four analyzers discussed. cyte counting.
Overview 4. Explain the derivation of calculated or indirectly mea- 8. Identify sources of error in automated cell counting
Coulter Instrumentation sured hemogram parameters for the same four and determine appropriate corrective action.
Sysmex Instrumentation analyzers.
Abbott Instrumentation
5. Explain the derivation of the white blood cell (WBC)
Siemens Healthcare
differential count on the different instruments
Diagnostics
Instrumentation discussed.
Automated Reticulocyte
Counting
Limitations and
Interferences
Calibration
Instrument Limitations
Sample Limitations
Clinical Utility of
Automated Hematology
Instrumentation

A
utomation provides greater accuracy and greater preci-
GENERAL PRINCIPLES OF
sion than manual methods. Since the 1980s, instru-
HEMATOLOGY INSTRUMENTATION
mentation has virtually replaced manual cell counting,
with the possible exception of phase platelet counting in Despite the number of hematology analyzers available from
certain circumstances. Hematology analyzers are marketed different manufacturers and their varying levels of sophistica-
by multiple instrument manufacturers. These analyzers typi- tion and complexity, most rely on only two basic principles
cally provide the eight standard hematology parameters of operation: electronic impedance (resistance) and optical
(complete blood count [CBC]) plus a three-part or five-part scatter. Electronic impedance, or low-voltage direct current (DC)
differential leukocyte count in less than 1 minute on 200L resistance, was developed by Coulter in the 1950s1,2 and is
of whole blood. Automation allows more efficient workload the most common methodology used. Radiofrequency (RF), or
management and more timely diagnosis and treatment of alternating current resistance, is a modification sometimes
disease. used in conjunction with DC electronic impedance. Technicon

*Deanne H. Chapman (1951-2007) made significant contributions to both the second and third editions of this chapter. The current authors wish
to remember Deanne as a respected colleague and friend.

598
CHAPTER 39 Automated Cell-Counting Instrumentation 599

Vacuum (6" Hg) Oscilloscope

Aperture current
100 fL
98 fL
96 fL
Internal 94 fL
electrode 92 fL
+ 90 fL
88 fL
External 86 fL
84 fL
electrode Blood cell 82 fL
suspension 80 fL
78 fL

Histogram

Aperture Aperture bath Aperture tube


Figure 39-1 Coulter principle of cell counting. (From Coulter Electronics:
Coulter STKR product reference manual, PN 4235547E, Hialeah, Fla, 1988,
Coulter Electronics.)

Instruments Corporation (Tarrytown, N.Y.) introduced dark-


field optical scanning in the 1960s, and Ortho Clinical Diag-
Relative number

nostics, Inc. (Raritan, N.J.), followed with a laser-based optical


instrument in the 1970s.3 Optical scatter, using both laser and
non-laser light, is frequently employed in todays hematology
instrumentation.
78 80 82 84 86 88 90 92 94 96 98 100 102
Electronic Impedance Femtoliters
The impedance principle of cell counting is based on the detec-
Figure 39-2 Oscilloscope display and histogram showing the construc-
tion and measurement of changes in electrical resistance pro-
tion of a frequency distribution graph. (Modified from Coulter Electronics:
duced by cells as they traverse a small aperture. Cells suspended
Significant advances in hematology: hematology education series, PN
in an electrically conductive diluent such as saline are pulled 4206115A, Hialeah, Fla, 1983, Coulter Electronics.)
through an aperture (orifice) in a glass tube. In the counting
chamber, or transducer assembly, low-frequency electrical
current is applied between an external electrode (suspended in enumerated between the lower and upper set thresholds for
the cell dilution) and an internal electrode (housed inside the each population. Size distribution histograms may be used for
aperture tube). Electrical resistance between the two electrodes, the evaluation of one cell population or subgroups within a
or impedance in the current, occurs as the cells pass through the population.5 The use of proprietary lytic reagents to control
sensing aperture, causing voltage pulses that are measurable shrinkage and lysis of specific cell types, as in the older Coulter
(Figure 39-1).4,5 Oscilloscope screens on some instruments S-Plus IV, STKR, and Sysmex E-5000 models, allows separation
display the pulses that are generated by the cells as they inter- and quantitation of white blood cells (WBCs) into three popu-
rupt the current. The number of pulses is proportional to the lations (lymphocytes, mononuclear cells, and granulocytes) for
number of cells counted. The size of the voltage pulse is directly the three-part differential on one size distribution histogram.6-8
proportional to the size (volume) of the cell, which allows dis- Several factors may affect size or volume measurements
crimination and counting of cells of specific sizes through the in impedance or volume displacement instruments. Aperture
use of threshold circuits. Pulses are collected and sorted (chan- diameter is crucial, and the red blood cell (RBC)/platelet aper-
nelized) according to their amplitude by pulse height analyzers. ture is smaller than the WBC aperture to increase platelet count-
The data are plotted on a frequency distribution graph, or size ing sensitivity. On earlier systems, protein buildup occurred,
distribution histogram, with relative number on the y-axis and decreasing the diameter of the orifice, slowing the flow of
size (channel number equivalent to a specific size) on the x-axis. cells, and increasing their relative electrical resistance. Protein
The histogram produced depicts the volume distribution of the buildup results in lower cell counts, which result in falsely
cells counted. Figure 39-2 illustrates the construction of a fre- elevated cell volumes. Impedance instruments once required
quency distribution graph. Size thresholds separate the cell frequent manual aperture cleaning, but current instruments
populations on the histogram, and the count is the cells incorporate burn circuits or other internal cleaning systems
600 PART VII Cell-Counting Automation

to prevent or slow protein buildup.6-9 Carryover of cells from Radiofrequency current


one sample to the next also is minimized by these internal
cleaning systems. Coincident passage of more than one cell at Direct current
a time through the orifice causes artificially large pulses, which
results in falsely increased cell volumes and falsely decreased DC supply
cell counts. This count reduction, or coincident passage loss, Detection
is statistically predictable (and mathematically correctable) chamber
Radiofrequency
because of its direct relationship to cell concentration and the External supply
size or effective volume of the aperture.7-9 Coincidence correc- electrode (+) Resistance
tion typically is completed by the analyzer computer before Electrolyte Condenser
final printout of cell counts from the instrument. Other factors solution
affecting pulse height include orientation of the cell in the
center of the aperture and deformability of the RBC, which may
be altered by decreased hemoglobin (Hb) content.10,11 Recircu- Aperture Internal electrode ()
lation of cells back into the sensing zone creates erroneous Figure 39-3 Radiofrequency/direct current (RF/DC) detection method,
pulses and falsely elevates cell counts. A backwash or sweep- showing simultaneous use of DC and RF in one measurement system on the
flow mechanism has been added to prevent recirculation of Sysmex SE-9500. (From TOA Medical Electronics Company: Sysmex
cells back into the sensing zone, and anomalously shaped SE-9500 operators manual [CN 461-2464-2], Kobe, Japan, 1997, TOA
pulses are edited out electronically.6,7,9 Medical Electronics Co.)
The use of hydrodynamic focusing avoids many of the
potential problems inherent in a rigid aperture system. The and basophils). DC and RF detection are two methods used by
sample stream is surrounded by a sheath fluid as it passes the Sysmex analyzers to perform WBC differentials.15,16
through the central axis of the aperture. Laminar flow allows
the central sample stream to narrow sufficiently to separate and Optical Scatter
align the cells into single file for passage through the sensing Optical scatter may be used as the primary methodology or in
zone.12-14 The outer sheath fluid minimizes protein buildup combination with other methods. In optical scatter systems
and plugs, eliminates recirculation of cells back into the sensing (flow cytometers), a hydrodynamically focused sample stream
zone with generation of spurious pulses, and reduces pulse is directed through a quartz flow cell past a focused light source
height irregularity because off-center cell passage is prevented (see Figure 33-3). The light source is generally a tungsten-
and better resolution of the blood cells is obtained. Coincident halogen lamp or a helium-neon laser (light amplification by
passage loss also is reduced because blood cells line up one stimulated emission of radiation). Laser light, termed mono-
after another in the direction of the flow.15 Laminar flow and chromatic light because it is emitted at a single wavelength,
hydrodynamic focusing are discussed further in Chapter 33. differs from brightfield light in its intensity, its coherence (i.e.,
it travels in phase), and its low divergence or spread. These
Radiofrequency characteristics allow for the detection of interference in the
Low-voltage DC impedance, as described previously, may be laser beam and enable enumeration and differentiation of cell
used in conjunction with RF resistance, or resistance to a high- types.12,17 Optical scatter may be used to study RBCs, WBCs,
voltage electromagnetic current flowing between both elec- and platelets.
trodes simultaneously. Although the total volume of the cell is As the cells pass through the sensing zone and interrupt the
proportional to the change in DC, the cell interior density (e.g., beam, light is scattered in all directions. Light scatter results
nuclear volume) is proportional to pulse size or change in the from the interaction between the processes of absorption, dif-
RF signal. Conductivity, as measured by this high-frequency fraction (bending around corners or the surface of a cell),
electromagnetic probe, is attenuated by nucleus-to-cytoplasm refraction (bending because of a change in speed), and reflec-
ratio, nuclear density, and cytoplasmic granulation. DC and RF tion (backward scatter of rays caused by an obstruction).18 The
voltage changes may be detected simultaneously and separated detection of scattered rays and their conversion into electrical
by two different pulse processing circuits.15,16 Figure 39-3 illus- signals is accomplished by photodetectors (photodiodes and
trates the simultaneous use of DC and RF current. photomultiplier tubes) at specific angles. Lenses fitted with
Two different cell properties, such as impedance and con- blocker bars to prevent nonscattered light from entering the
ductivity, can be plotted against each other to create a two- detector are used to collect the scattered light. A series of filters
dimensional distribution cytogram or scatterplot (Figure 39-4). and mirrors separate the varying wavelengths and present them
Such plots display the cell populations as clusters, with the to the photodetectors. Photodiodes convert light photons to
number of dots in each cluster representing the concentration electronic signals proportional in magnitude to the amount of
of that cell type. Computer cluster analysis can determine abso- light collected. Photomultiplier tubes are used to collect the
lute counts for specific cell populations. The use of multiple weaker signals produced at a 90-degree angle and multiply the
methods by a given instrument for the determination of at least photoelectrons into stronger, useful signals. Analogue-to-
two cell properties allows the separation of WBCs into a five-part digital converters change the electronic pulses to digital signals
differential (neutrophils, lymphocytes, monocytes, eosinophils, for computer analysis.12,17
CHAPTER 39 Automated Cell-Counting Instrumentation 601

RF D I F F

RF signal DC

DC signal
Figure 39-4 Illustration of cell size/volume measurement with direct current (DC) voltage change versus measurement of cell nuclear volume/complexity
with change in the radiofrequency (RF) signal. The two measurements can be plotted against each other to form a two-dimensional distribution scatterplot.
(From TOA Medical Electronics Company: Sysmex SE-9500 operators manual [CN 461-2464-2], Kobe, Japan, 1997, TOA Medical Electronics Co.)

Forward-angle light scatter (0 degrees) correlates with cell Healthcare Diagnostics, Inc. (Deerfield, IL),27 Beckman Coulter,
volume or size, primarily because of diffraction of light. Inc. (Brea, Calif.),28 and Sysmex Corporation (Kobe, Japan).29
Orthogonal light scatter (90 degrees), or side scatter, results The following discussion is limited to instrumentation pro-
from refraction and reflection of light from larger structures duced by four of these suppliers. Emphasis is not placed on
inside the cell and correlates with degree of internal complex- sample size or handling, speed, level of automation, or com-
ity. Forward low-angle scatter (2 to 3 degrees) and forward parison of instruments or manufacturers. Likewise, technology
high-angle scatter (5 to 15 degrees) also correlate with cell continues to improve, and the newest (or most recent) models
volume and refractive index or with internal complexity.17,19,20 produced by a manufacturer may not be mentioned. Instead,
Differential scatter is the combination of this low-angle and a detailed description of primary methods used by these manu-
high-angle forward light scatter and is primarily used on facturers is given to show the application of, and clarify further,
Siemens systems for cellular analysis. The angles of light scatter the principles presented earlier and to enable the technologist
measured by the different flow cytometers are manufacturer better to interpret patient data, including instrument-generated
and method specific. histograms and cytograms, in the hematology laboratory.
Scatter properties at different angles may be plotted against Table 39-1 summarizes methods used for hemogram determi-
each other to generate two-dimensional cytograms or scatter- nation on four major hematology instruments. Table 39-2
plots, as on the Abbott CELL-DYN instruments.21,22 Optical summarizes methods used for WBC differential determination
scatter may also be plotted against absorption, as on the on the same four analyzers.
Siemens systems,23,24 or against volume, as on the larger Coulter Hematology analyzers have some common basic compo-
systems.9 Computer cluster analysis of the cytograms may yield nents, including hydraulics, pneumatics, and electrical systems.
quantitative and qualitative information. The hydraulics system includes aspirating unit, dispensers,
diluters, mixing chambers, aperture baths or flow cells or both,
and a hemoglobinometer. The pneumatics system includes
PRINCIPAL CELL-COUNTING INSTRUMENTS
vacuums and pressures required for operating the valves and
Overview moving the sample through the hydraulics system. The electri-
Hematology analyzers are produced by multiple manu cal system controls operational sequences of the total system
facturers, including, but not limited to, Abbott Laboratories and includes electronic analyzers and computing circuitry for
(Abbott Park, Ill.),25 HORIBA Medical (Irvine, Calif.),26 Siemens processing the data generated. Some older-model instruments
602 PART VII Cell-Counting Automation

TABLE 39-1 Methods for Hemogram and Reticulocyte Count Determination on Four Major Hematology Instruments
Parameter Coulter LH 750 Sysmex XE-2100 Abbott CELL-DYN 4000 Siemens ADVIA 2120
WBC Impedance, hydrodynamic Hydrodynamic focusing, DC Optical scatter (primary count), Hydrodynamic focusing, optical
focusing detection (impedance) impedance (secondary count) scatter and absorption
RBC Impedance Hydrodynamic focusing, DC Impedance Hydrodynamic focusing, laser
detection (impedance) low-angle (2-3 degree) and
high-angle (5-15 degree) scatter
Hb Modified cyanmethemoglobin SLS-Hb (555nm) Modified cyanmethemoglobin Modified cyanmethemoglobin
(525nm) (540nm) (546nm)
Hct (RBC MCV)/10 Cumulative pulse height (RBC MCV)/10 (RBC MCV)/10
detection
MCV Mean of RBC volume (Hct/RBC) 10 Mean of RBC volume Mean of RBC volume histogram
distribution histogram distribution histogram
MCH (Hb/RBC) 10 (Hb/RBC) 10 (Hb/RBC) 10 (Hb/RBC) 10
MCHC (Hb/Hct) 100 (Hb/Hct) 100 (Hb/Hct) 100 (Hb/Hct) 100
Platelet count Impedance (2-20 fL): Hydrodynamic focusing, DC Impedance (approximately Hydrodynamic focusing, laser
least-squares fit of detection (impedance) 2-30 fL) low-angle (2-3 degree) and
volume distribution (approximately 2-30 fL) high-angle (5-15 degree) scatter
histogram (0-70 fL) (1-60 fL)
RDW CV (%) of RBC histogram: RDW SD (fL) or RDW CV Relative value, equivalent to CV CV (%) of RBC histogram: (SD/
(SD/MCV) 100 (%) available MCV) 100
Reticulocyte Supravital staining (new Supravital staining (auramine Proprietary stain (CD4K530), Supravital staining (oxazine 750);
count methylene blue); volume, O); fluorescence detection multiangle scatter, and low-angle (2-3 degree) and
conductivity, optical fluorescence detection high-angle (5-15 degree) optical
scatter (VCS technology) scatter and absorbance

CV, Coefficient of variation; DC, direct current; Hb, hemoglobin; Hct, hematocrit; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration; MCV, mean cell
volume; RBC, red blood cell (or count); RDW, RBC distribution width; SD, standard deviation; SLS, sodium lauryl sulfate; VCS, volume, conductivity, scatter; WBC, white blood
cell (or count).

TABLE 39-2 Methods for White Blood Cell Differential Determination on Four Major Hematology Instruments
Coulter Abbott
Parameter LH 750 Sysmex XE-2100 CELL-DYN 4000 Siemens ADVIA 2120
Neutrophils VCS Optical scatter and fluorescent staining MAPSS Peroxidase staining, optical scatter and absorption
Lymphocytes VCS Optical scatter and fluorescent staining MAPSS Peroxidase staining, optical scatter and absorption
Monocytes VCS Optical scatter and fluorescent staining MAPSS Peroxidase staining, optical scatter and absorption
Eosinophils VCS Optical scatter and fluorescent staining MAPSS Peroxidase staining, optical scatter and absorption
Basophils VCS Optical scatter and fluorescent staining MAPSS Differential lysis, laser low-angle (2-3 degree) and
high-angle (5-15 degree) scatter

MAPSS, Multiangle polarized scatter separation; VCS, volume, conductivity, scatter.

have oscilloscope screens that display the electrical pulses in and storage of quality control files; patient data storage and
real time as the cells are counted. A data display unit receives retrieval, with checks, panic or critical value flagging, and
information from the analyzer and prints results, histograms, automatic verification of patient results based on user-defined
or cytograms. algorithms; host query with the laboratory or hospital informa-
Sample handling varies from instrument to instrument tion system to allow random access discrete testing capability;
based on degree of automation, and systems range from dis- analysis of animal specimens; and even analysis of body fluids.
crete analyzers to walkaway systems with front-end load
capability. Computer functions also vary, with the larger instru- Coulter Instrumentation
ments having extensive microprocessor and data management Beckman Coulter, Inc., manufactures an extensive line of
capabilities. Computer software capabilities include automatic hematology analyzers, including the smaller ONYX series that
start-up and shutdown, with internal diagnostic self-checks provide complete RBC, platelet, and WBC analysis with three-
and some maintenance; quality control, with automatic review part differential; the larger MAXM and STKS systems that
of quality control data, calculations, graphs, moving averages, perform a CBC with five-part differential and reticulocyte
CHAPTER 39 Automated Cell-Counting Instrumentation 603

analysis using off-line sample preparation; and the GEN-S and such as debris, and in the larger region, such as small RBCs.
newer LH 780 systems, part of the LH 700 series, that provide The mean platelet volume (MPV), analogous to the RBC MCV,
a fully automated on-line reticulocyte analysis.28 The LH series also is derived from the platelet histogram. Reference values
also has the capability to perform CD4 and CD8 counts.30-32 for the MPV are about 7.8 to 11.0fL. The MPV increases slightly
Coulter instruments typically have two measurement channels as the sample sits in the anticoagulant, ethylenediaminetet-
in the hydraulics system for determining the hemogram data. raacetic acid (EDTA).5
The RBC and WBC counts and hemoglobin are considered to Many older-model Coulter instruments, such as the STKR,
be measured directly. The aspirated whole-blood sample is and the newer smaller models, such as the ONYX, provide
divided into two aliquots, and each is mixed with an isotonic three-part leukocyte subpopulation analysis, which differenti-
diluent. The first dilution is delivered to the RBC aperture ates WBCs into lymphocytes, mononuclear cells, and granulo-
chamber, and the second is delivered to the WBC aperture cytes. In the WBC channel, a special lysing reagent causes
chamber. In the RBC chamber, RBCs and platelets are counted differential shrinkage of the leukocytes, which allows the differ-
and discriminated by electrical impedance as the cells are ent cells to be counted and sized based on their impedance. A
pulled through each of three sensing apertures (50m in WBC histogram is constructed from the channelized data.
diameter, 60m in length). Particles 2 to 20fL are counted as Particles between approximately 35 and 90fL are considered
platelets, and particles greater than 36fL are counted as RBCs. lymphocytes; particles between 90 and 160fL, mononucle-
In the WBC chamber, a reagent to lyse RBCs and release hemo- ars (monocytes, blasts, immature granulocytes, and reactive
globin is added before WBCs are counted simultaneously by [atypical] lymphocytes); and particles between 160 and
impedance in each of three sensing apertures (100m in 450fL, granulocytes. This allows the calculation of relative and
diameter, 75m in length). Alternatively, some models employ absolute numbers for these three populations.6 Proprietary
consecutive counts in the same RBC or WBC aperture. After computerized algorithms further allow flagging for increased
counting cycles are completed, the WBC dilution is passed eosinophils or basophils or both and interpretation of the
to the hemoglobinometer for determination of hemoglobin histogram differential, including flagging for abnormal cells,
concentration (light transmittance read at a wavelength of such as reactive lymphocytes and blasts.7 When cell popula-
525nm). Electrical pulses generated in the counting cycles are tions overlap or a distinct separation of populations does not
sent to the analyzer for editing, coincidence correction, and exist, a region alarm (R flag) may be triggered that indicates
digital conversion. Two of the three counts obtained in the RBC the area of interference on the size-distribution histogram. An
and the WBC baths must match within specified limits for the R1 flag represents excess signals at the lower threshold region
counts to be accepted by the instrument.5,9 This multiple count- of the WBC histogram and a questionable WBC count. This
ing procedure prevents data errors resulting from aperture interference is visualized as a high takeoff of the curve and
obstructions or statistical outliers and allows for excellent may indicate the presence of nucleated RBCs, clumped plate-
reproducibility on the Coulter instruments. lets, unlysed RBCs, or electronic noise.6,7 Figure 39-5 shows
Pulse height is measured and categorized by pulse height composite printouts from the Coulter STKR that include the
analyzers; 256 channels are used for WBC and RBC analysis, interpretive differential.
and 64 channels are used for platelet analysis. Size-distribution More recent Coulter instruments, the STKS, MAXM, GEN-S,
histograms of WBC, RBC, and platelet populations are gener- and LH 750, generate hemogram data (including the WBC
ated. The RBC mean cell volume (MCV) is the average volume count) exactly as before but use Coulters proprietary VCS
of the RBCs taken from the size distribution data. The hema- (volume, conductivity, scatter) technology in a separate channel to
tocrit (Hct), mean cell hemoglobin (MCH), and mean cell evaluate WBCs for the determination of a five-part differential.
hemoglobin concentration (MCHC) are calculated from mea- The VCS technology includes the volumetric sizing of cells by
sured and derived values. The RBC distribution width (RDW) impedance, conductivity measurements of cells, and laser light
is calculated directly from the histogram as the coefficient of scatter, all performed simultaneously for each cell. After RBCs
variation (CV) of the RBC volume distribution, with a reference are lysed and WBCs are treated with a stabilizing reagent to
range of 11.5% to 14.5%.5 The RDW is an index of anisocyto- maintain them in a near-native state, a hydrodynamically
sis, but may be falsely skewed because it reflects the ratio of focused sample stream is directed through the flow cell past
the standard deviation (SD) to MCV. That is, an RBC distribu- the sensing zone. Low-frequency DC measures size, whereas a
tion histogram with normal divergence, but with a decreased high-frequency electromagnetic probe measures conductivity,
MCV may imply a high RDW, falsely indicating increased an indicator of cellular internal content. The conductivity
anisocytosis. MCV and RDW are used by the instrument to flag signal is corrected for cellular volume, which yields a unique
possible anisocytosis, microcytosis, and macrocytosis.9 measurement called opacity. Each cell also is scanned with
Platelets are counted within the range of 2 to 20fL, and a monochromatic laser light that reveals information about the
size-distribution histogram is constructed. If the platelet size cell surface, such as structure, shape, and reflectivity. Coulters
distribution meets specified criteria, a statistical least-squares unique rotated light scatter detection method, which covers a
method is applied to the raw data to fit the data to a log-normal 10-degree to 70-degree range, allows for separation of cells
curve. The curve is extrapolated from 0 to 70fL, and the final with similar size, but different scatter characteristics. More than
count is derived from this extended curve. This fitting proce- 8000 WBCs are analyzed in each sample.31,33 Beckman Coul-
dure eliminates interference from particles in the noise region, ters newest analyzer, the UniCel DxH 800, utilizes the volume
604 PART VII Cell-Counting Automation

A B

Figure 39-5 Printouts from the Coulter STKR showing the interpretive differential. A, Note the three distinct white blood cell (WBC) populations, normal
gaussian or bell-shaped distribution of red blood cells (RBCs), and right-skewed or log-normal distribution of platelets. B, Note the left shift in the WBC his-
togram with possible interference at the lower threshold region. R2 flag indicates interference and loss of valley owing to overlap or insufficient separation
between the lymphocyte and mononuclear populations at the 90-fL region. RM flag indicates interference at more than one region. Eosinophil data have been
suppressed. Also note the abnormal platelet size distribution with a low platelet count. Manual 200-cell differential counts on the same specimens: A, 42.5%
neutrophils (37% segmented neutrophils, 5.5% bands), 41.5% lymphocytes, 4.5% monocytes, 1% basophils, 0.5% metamyelocytes, 9.5% myelocytes, 0.5%
reactive lymphocytes; B, 51% neutrophils (23% segmented neutrophils, 28% bands), 12% lymphocytes, 9.5% monocytes, 1% metamyelocytes, 1.5% myelo-
cytes, 25% reactive lymphocytes, and 17 nucleated RBCs/100 WBCs.

and conductivity as well as low angle scatter as in earlier distributional abnormalities, such as eosinophilia or lym
models but also measures additional lower and higher median phocytopenia (based on absolute eosinophil or lymphocyte
angle scatter to provide additional information for cell identi- counts); and (2) instrument specific, primarily suspect flags
fication and flagging.34 for morphologic abnormalities. For distributional flags, the
This combination of technologies provides a three- user establishes reference ranges and programs the instrument
dimensional plot or cytograph of the WBC populations, which to flag each parameter as high or low. Suspect flags indicating
are separated by computer cluster analysis. Two-dimensional the possible presence of abnormal cells are triggered when
scatterplots of the three measurements represent different cell populations fall outside expected regions or when
views of the cytograph. The discriminate function 1 (DF 1) specific statistical limitations are exceeded. Instrument-specific
scatterplot of volume (y-axis) versus light scatter (x-axis) shows suspect flags on the Coulter LH 700 series include immature
clear separation of lymphocytes, monocytes, neutrophils, and granulocytes/bands, blasts, variant lymphocytes, nucleated
eosinophils. Basophils are hidden behind the lymphocytes in RBCs, and platelet clumps. Inadequate separation of cell popu-
this scatterplot but are separated by conductivity owing to their lations may disallow reporting of differential results by the
cytoplasmic granulation. The DF 2 scatterplot of volume instrument and may elicit a review slide message.9,35
(y-axis) versus conductivity (x-axis) shows lymphocytes, mono- On the LH 700 series, Coulter has added an IntelliKinetics
cytes, and neutrophils as the prominent populations. When the application. This application is used to ensure consistency
neutrophil population is removed from the DF 2 scatterplot, with the kinetic reactions. It provides the instrument the best
the basophil population may be visualized on the z-axis (DF signals for analysis independent of laboratory environment
3 display). DF 2 and DF 3 scatterplots are available at operator variations. Compared with earlier models Coulter IntelliKinet-
request. Single-parameter histograms of volume, conductivity, ics provides better separation of cell populations for WBCs and
and light scatter also are available.9,33 Figure 39-6 represents a reticulocytes, which enables better analysis by the system
standard patient printout from the Coulter LH 750 showing a algorithms.35
DF 1 scatterplot.
Two types of WBC flags (alarms or indicators of abnormal- Sysmex Instrumentation
ity) are generated on all hematology analyzers that provide a Sysmex Corporation, formerly TOA Medical Electronics
WBC differential count: (1) user defined, primarily set for Company, Ltd., manufactures a full line of hematology
CHAPTER 39 Automated Cell-Counting Instrumentation 605

Coulter LH 750

Date: 5/26/2005 Sample ID: 4004 Cass / Pos: 000904 Operator ID: LABADMIN
Time: 11:28:26 Sample Type: CD A NO Read Listname: 37H5QDD4 Instrument: LH1N

Suspect Definitive
WBC 8.8 103/uL WBC Histogram
NE % 65.7 %
LY % 25.7 %
MO % 7.9 %
EO % 0.5 L % 90 100 200 300 fL
BA % 0.2 L %
NRBC % 0.0 %
NE # 5.8 103/L
A
LY # 2.3 103/L
MO # 0.7 103/L
EO # 0.0 L 103/L
BA # 0.0 103/L
NRBC # 0.0 103/L

RBC 4.24 L 106/L


HGB 12.4 L g/dL
HCT 37.3 L % B RBC Histogram
MCV 88.0 fL
MCH 29.2 pg
MCHC 33.1 g/dL
RDW 14.7 H %
50 100 200 300 fL

C PLT Histogram
PLT 344 103/L
MPV 7.2 fL

5 10 20 30 fL

Figure 39-6 Composite scatterplots/histograms obtained from the Coulter LH 750 for the same normal specimen analyzed in Figures 39-7, 39-9, and
39-12. A, White blood cell (WBC) two-dimensional scatterplot shows volume as determined by impedance on the y-axis versus light scatter (discriminate
function 1, or DF 1) on the x-axis. Computer-generated floating discriminators separate lymphocyte (LY), monocyte (MO), neutrophil (NE), and eosinophil (EO)
regions. Data below the WBC threshold represent non-WBC debris. Large or clumped platelets, nucleated red blood cells (RBCs), WBC fragments, and unlysed
RBCs in abnormal samples should fall below this threshold. DF 2 and DF 3 scatterplots (not shown) depict volume (y-axis) versus conductivity (x-axis). Basophils
(BA) are best visualized on the DF 3 display, from which the neutrophil population has been removed. All two-dimensional scatterplots, single-parameter
histograms, and the three-dimensional cytograph may be reviewed at operator request. The relative value (percent) of each of the five WBC populations derived
from this three-dimensional analysis is multiplied by the WBC count derived in a separate impedance channel for determination of the absolute counts (number).
B and C, Relative number (y-axis) versus volume distribution (x-axis) histograms for RBCs and platelets (PLT). Note the normal gaussian or bell-shaped dis-
tribution of the RBCs in B and the right-skewed or log-normal distribution of platelets in C. The small population of larger RBCs on the right of the RBC curve
represents coincident passage of doublets or triplets or both through the impedance aperture, which creates larger pulses. This portion of the RBC curve is
excluded from the calculation of the red cell distribution width (RDW). The mean platelet volume (MPV) is analogous to the mean cell volume (MCV).

analyzers, including the small KX-21N and K-4500 that provide chambers, diluted WBC and RBC samples are aspirated through
complete RBC, platelet, and WBC analysis with three-part dif- the different apertures and counted using the impedance (DC
ferential; the larger XT-1800i (SF-3000 and SE-9000) that per- detection) method for counting and sizing cells. Two unique
forms a CBC with five-part differential; and the newer XE-5000 features enhance the impedance technology: (1) in the RBC/
series that also provides a fully automated reticulocyte platelet channel, a sheathed stream with hydrodynamic focus-
count.29,36,37 The WBC, RBC, hemoglobin, hematocrit, and ing is used to direct cells through the aperture, which reduces
platelet counts are considered to be measured directly. Three coincident passage, particle size distortion, and recirculation of
hydraulic subsystems are used for determining the hemogram: blood cells around the aperture; and (2) in the WBC and RBC/
the WBC channel, the RBC/platelet channel, and a separate platelet channels, floating thresholds are used to discrimi-
hemoglobin channel. In the WBC and RBC transducer nate each cell population.8,15,16
606 PART VII Cell-Counting Automation

As cells pass through the apertures, signals are transmitted of RF detection signals (y-axis) versus DC detection signals
in sequence to the analogue circuit and particle size distribu- (x-axis) allows separation of the WBCs into lymphocytes,
tion analysis circuits for conversion to cumulative cell size monocytes, and granulocytes. Floating discriminators deter-
distribution data. Particle size distribution curves are con- mine the optimal separation between these populations. Gran-
structed, and optimal position of the autodiscrimination level ulocytes are analyzed further in the IMI (immature myeloid
(i.e., threshold) is set by the microprocessor for each cell popu- information) detection chamber to determine immature
lation. The lower platelet threshold is automatically adjusted (myeloid) information. RBCs are lysed, and WBCs other than
in the 2- to 6-fL volume range, and the upper threshold is immature granulocytes are selectively shrunk by temperature
adjusted in the 12- to 30-fL range, based on particle volume and chemically controlled reactions. Analysis of the treated
distribution. Likewise, the RBC lower and upper thresholds sample using the DC/RF detection method allows separation
may be set in the 25- to 75-fL and 200- to 250-fL volume of immature cells on the IMI scattergram. A similar differential
ranges. This floating threshold circuitry allows for discrimina- shrinkage and lysis method is also used in the EO and BASO
tion of cell populations on a sample-by-sample basis. Cell chambers. That is, eosinophils and basophils are counted by
counts are based on pulses between the lower and upper impedance (DC detection) in separate chambers in which the
autodiscriminator levels, with dilution ratio, volume counted, RBCs are lysed and WBCs other than eosinophils or basophils
and coincident passage error accounted for in the final are selectively shrunk by temperature and chemically con-
computer-generated numbers. In the RBC channel, the floating trolled reactions. Eosinophils and basophils are subtracted
discriminator is particularly useful in separating platelets from the granulocyte count derived from the DIFF scattergram
from small RBCs. The hematocrit also is determined from analysis to determine the neutrophil count. User-defined dis-
the RBC/platelet channel, based on the principle that pulse tributional flags may be set, and instrument-specific suspect
height generated by the RBC is proportional to cell volume. flags, similar to those described for the Coulter LH 700 series,
The hematocrit is the RBC cumulative pulse height and is are triggered for the possible presence of morphologic abnor-
considered a true relative percentage volume of erythrocytes.8,15 malities.15,16 A POSITIVE or NEGATIVE interpretive message is
In the hemoglobin flow cell, hemoglobin is converted to displayed. Figure 39-7 shows a patient report from the XE-2100
oxyhemoglobin, which combines with sodium lauryl sulfate analyzing the same patient sample for which data are given in
to become a sodium lauryl sulfatehemoglobin hemachrome Figure 39-6.
molecule, with concentration measured as absorbance at
555nm.16,38 Abbott Instrumentation
The following indices are calculated in the microprocessor Instruments offered by Abbott Laboratories include the smaller
using directly measured or derived parameters: MCV, MCH, CELL-DYN Emerald, which provides complete RBC, platelet,
MCHC, RDW-SD, RDW-CV, MPV, and plateletcrit. RDW-SD is and WBC analysis with three-part differential; and the larger
the RBC arithmetic distribution width measured at 20% of the CELL-DYN Sapphire and the mid range CELL-DYN Ruby, both
height of the RBC curve, reported in femtoliters, with a refer- of which provide a CBC with five-part differential and random
ence interval of 37 to 54fL. RDW-CV is the RDW reported as fully automated reticulocyte analysis.41 The CELL-DYN 4000
a CV. Plateletcrit is the platelet volume ratio, analogous to the system has three independent measurement channels for deter-
hematocrit. MPV is calculated from the plateletcrit and platelet mining the hemogram and differential: (1) an optical channel
count just as erythrocyte MCV is calculated from the hematocrit for WBC count and differential data, (2) an impedance channel
and RBC count. The proportion of platelets greater than 12fL for RBC and platelet data, and (3) a hemoglobin channel for
in the total platelet count may be an indicator of possible hemoglobin determination.21,22 An additional impedance
platelet clumping, giant platelets, or cell fragments.8,15,16 The channel for determining a WBC impedance count is used as an
XE series has the capability to run the platelet counting in the internal check of the primary WBC optical count. When the
optical mode, which eliminates common interferences found WBC impedance count and WBC optical count differ, an algo-
with impedance counting. In the optical mode, a new param- rithm selects the most appropriate value for the reported
eter, immature platelet fraction or IPF, can be measured to WBC.22 The WBC, RBC, hemoglobin, and platelet parameters
provide additional information concerning platelet kinetics in are considered to be measured directly. A 60- to 70-m aper-
cases of thromobocytopenia.39,40 ture is used in the RBC/platelet transducer assembly for count-
The SE-9000/9500 uses four detection chambers to analyze ing and sizing of RBCs and platelets by the electronic impedance
WBCs and obtain a five-part differential: the DIFF, IMI, EO, method.
and BASO chambers. The high-end instrumention such as the A unique von Behrens plate is located in the RBC/platelet
XE-5000 in the XE series has a six-part differential: neutrophils, counting chamber to minimize the effect of recirculating cells.
lymphocytes, monocytes, eosinophils, basophils, and imma- Pulses are collected and sorted in 256 channels according to
ture granulocytes. Every differential performed generates a per- their amplitudes: particles between 1 and 35fL are included in
centage and absolute number for immature granulocytes, thus the initial platelet data, and particles greater than 35fL are
providing valuable information about the complete differen- counted as RBCs. Floating thresholds are used to determine the
tial.37 In the DIFF detection chamber, RBCs are hemolyzed and best separation of the platelet population and to eliminate
WBCs are analyzed simultaneously by low-frequency DC and interference, such as noise, debris, or small RBCs, from the
high-frequency current (DC/RF detection method). A scattergram count. Coincident passage loss is corrected for in the final RBC
CHAPTER 39 Automated Cell-Counting Instrumentation 607

SYSMEX XE 2100
Sample No.: ERR000000000005 Rack: 9 Tube: 4 05/25/2005 15:41:55
Patient ID: 4004 Ward: Dr.:
Name: Birth: Sex:
Comments: Inst.ID:FLO

Negative
A B
DIFF WBC/BASO

SFL

FSC
WBC 8.73 [103/L]
RBC 4.14 - [106/L]
HGB 12.2 [g/dL]
HCT 37.9 [%]
MCV 91.5 [fL]
MCH 29.5 [pg]
MCHC 32.2 [g/dL] C D
RET PLT-O
PLT 321 [103/L] FSC

FSC
RDW-SD 48.0 [fL]
RDW-CV 14.7 [%]
MPV 8.7 - [fL]
NEUT 5.55 [103/L] 63.6 [%]
LYMPH 2.00 [103/L] 22.9 [%]
MONO 1.12 + [103/L] 12.8 [%]
EO 0.04 [103/L] 0.5 [%]
E F
BASO 0.02 [103/L] 0.2 [%]
RET 1.27 [%] 0.0526 [106/uL] RBC PLT
IRF 11.4 [%]

Figure 39-7 Composite histograms/scatterplots obtained from the Sysmex XE-2100 for the same normal specimen analyzed in Figures 39-6, 39-9, and
39-12. In the DIFF scatterplot (A), high-frequency current (radiofrequency) detection signals on the y-axis are plotted against direct current (DC) detection
signals on the x-axis. Floating discriminators determine optimal separation of lymphocytes (LYMPH), monocytes (MONO), and neutrophils (NEUT). For the WBC/
BASO scatterplot (B), the total number of white blood cells (WBCs) is counted optically and a second count is performed, yielding the total number of basophils
(BASOs) in the WBC count, as illustrated. BASO counts are determined by enumerating cells between the autodiscriminator level and an upper fixed discrimi-
nator position. The eosinophil (EO) distribution histogram is not shown. The absolute neutrophil count (NEUT) ( 103/L) is computed by subtracting the EO
and BASO counts from the 103 result derived from the scatterplot analysis. Relative differential results (percentages) are computed by dividing absolute
numbers by total WBC count. C, The reticulocyte channel performs an optical count and measures mature red blood cells (RBCs), optical platelets, and reticu-
locytes, yielding a two-dimensional scatterplot analysis of the reticulocytes (RET) and the platelets. Lateral fluorescent light scatter on the x-axis is plotted
against forward light scatter on the y-axis. This analysis also yields the optical platelet scatterplot (PLT-O) as illustrated in D. E and F, Relative number (y-axis)
versus size distribution (x-axis) histograms for RBCs and platelets (PLT). Note the bell-shaped RBC curve in E with little interference on the right side from
coincident passage, minimized by hydrodynamic focusing in the RBC/PLT channel. The red cell distribution width, RDW (calculated in terms of both standard
deviation [RDW-SD] and coefficient of variation [RDW-CV]) is determined at the 20% relative height level to exclude any possible interference. FSC, Forward
scatter; SFL, side fluorescence.

and platelet counts. RBC pulse editing is applied before MCV hemiglobincyanide method that measures absorbance at
derivation to compensate for aberrant pulses produced by 540nm. Hematocrit, MCH, and MCHC are calculated from
nonaxial passage of RBCs through the aperture. The MCV is the the directly measured or derived parameters. The RDW, equiva-
average volume of the RBCs derived from RBC size distribution lent to CV, is a relative value, derived from the RBC histogram
data. Hemoglobin is measured directly using a modified by using the 20th and 80th percentiles. The platelet analysis is
608 PART VII Cell-Counting Automation

based on a two-dimensional optical platelet count using fluo- of eosinophils and neutrophils on this display. Dynamic
rescent technology, the same technology used for direct nucle- thresholds are used for best separation of the different popula-
ated RBC counting. Further analysis of platelets and platelet tions in the various scatterplots. Each cell type is identified with
aggregates can be performed by using platelet-specific a distinct color, so that after all classifications are made and
immunolabeling.42-44 Other indices available include MPV and size (0 degrees) is plotted against complexity (7 degrees), each
plateletcrit (analogous to hematocrit).21,22 cell population can be visualized easily by the operator on the
The WBC count and differential are derived from the optical data terminal screen. Other scatterplots (90 degrees/0 degrees,
channel using CELL-DYNs patented multiangle polarized 90 degrees depolarized/0 degrees, 90 degrees depolarized/7
scatter separation (MAPSS) technology. A hydrodynamically degrees) are available and may be displayed at operator request.
focused sample stream is directed through a quartz flow cell On earlier instruments, the 7-degree measurement for com-
past a focused light source, an argon ion laser. Scattered light plexity was referred to as the 10-degree value. The change
is measured at multiple angles: 0-degree forward light scatter reflects use of the midrange of the angle instead of the end
measurement is used for determination of cell size, 90-degree range; however, it still provides the same measurement and
orthogonal light scatter measurement is used for determina- information.46,47 As on the previously described instruments,
tion of cellular lobularity, 7-degree narrow-angle scatter mea- user-defined distributional flags may be set, and instrument-
surement is used to correlate with cellular complexity, and specific suspect flags may alert the operator to the presence of
90-degree depolarized light scatter measurement is used for abnormal cells.21,46 Figure 39-9 represents a patient printout
evaluation of cellular granularity. Orthogonal light scatter is from the CELL-DYN 3700.
split, with one portion directed to a 90-degree photomultiplier
tube and the other portion directed through a polarizer to the Siemens Healthcare
90-degree depolarized photomultiplier tube. Light that has Diagnostics Instrumentation
changed polarization (depolarized) is the only light that can Siemens Healthcare Diagnostics Inc. manufactures the ADVIA
be detected by the 90-degree depolarized photomultiplier 2120 and 2120i, the next generation of the ADVIA 120.27,48 The
tube. Various combinations of these four measurements are ADVIA 2120, 2120i, and 120 use much of the same technology
used to differentiate and quantify the five major WBC sub- as the older Technicon H-1, H-2, and H-3 systems.49 Siemens
populations: neutrophils, lymphocytes, monocytes, eosino- has simplified the hydraulics and operations of the analyzer by
phils, and basophils.21,45 Figure 39-8 illustrates Abbotts MAPSS replacing multiple complex hydraulic systems with a unified
technology. fluids circuit assembly, or Unifluidics technology. The ADVIA
The light scatter signals are converted into electrical signals, 2120, 2120i, and 120 provide a complete hemogram and WBC
sorted into 256 channels on the basis of amplitude for each differential while also providing a fully automated reticulocyte
angle of light measured, and graphically presented as scatter- count.23,24,48
plots. Scatter information from the different angles is plotted Four independent measurement channels are used in deter-
in various combinations: 90 degrees/7 degrees, or lobularity mining the hemogram and differential: (1) RBC/platelet
versus complexity; 0 degrees/7 degrees, size versus complexity; channel, (2) hemoglobin channel, and (3) peroxidase (PEROX)
and 90 degrees depolarized/90 degrees, granularity versus lob- and (4) basophil-lobularity (BASO) channels for WBC and
ularity. Lobularity (y-axis) plotted against complexity (x-axis) differential data. WBC, RBC, hemoglobin, and platelets are
yields separation of mononuclear and segmented (poly measured directly. Hemoglobin is determined using a modi-
morphonuclear neutrophil) subpopulations. (Basophils cluster fied cyanmethemoglobin method that measures absorbance in
with the mononuclears in this analysis, because the basophil a colorimeter flow cuvette at approximately 546nm. The RBC/
granules dissolve in the sheath reagent, and the degranulated platelet method uses flow cytometric light scattering measure-
basophil is a less complex cell.) Each cell in the two clusters ments determined as cells pass through a sheath-stream flow
is identified as a mononuclear or segmented neutrophil for cell past a laser optical assembly (laser diode light source).
further evaluation. RBCs and platelets are isovolumetrically sphered before enter-
The mononuclear subpopulation is plotted on a 0-degree/7- ing the flow cell to eliminate optical orientation noise. Laser
degree scatterplot, with size on the y-axis and complexity on light scattered at two different angular intervalslow angle (2
the x-axis. Three populations (lymphocytes, monocytes, and to 3 degrees), correlating with cell volume or size, and high
basophils) are seen clearly on this display. Nucleated RBCs, angle (5 to 15 degrees), correlating with internal complexity
unlysed RBCs, giant platelets, and platelet clumps fall below (i.e., refractive index or hemoglobin concentration)is mea-
the lymphocyte cluster on this scatterplot and are excluded sured simultaneously (Figure 39-10). This unique differential
from the WBC count and differential. Information from the scatter technique, in combination with isovolumetric spher-
WBC impedance channel also is used in discriminating these ing, eliminates the adverse effect of variation in cellular hemo-
particles.22 globin concentration on the determination of RBC volume
The segmented neutrophil subpopulation is plotted on a (as seen by differences in cellular deformability affecting the
90-degree depolarized/90-degree scatterplot, with granularity pulse height generated on impedance instruments).10,50 The
on the y-axis and lobularity on the x-axis. Because of the Mie theory of light scatter of dielectric spheres18 is applied
unique nature of eosinophil granules, eosinophils scatter more to plot scatter-intensity signals from the two angles against
90-degree depolarized light, which allows clear separation each other for a cell-by-cell RBC volume (y-axis) versus
CHAPTER 39 Automated Cell-Counting Instrumentation 609

B C D
Figure 39-8 A, Multiangle polarized scatter separation (MAPSS) technology. Cells are measured and characterized by looking at light scatter from four
different angles. B, Mononuclear and polymorphonuclear scatter with MAPPS technology. It plots the granularity/lobularity with the X axis at 90 degrees and
the Y axis at 10 degrees. Size and complexity are measured on the X axis at 10 degrees and the Y axis at 0 degrees. The system uses algorithms to further
separate the two populations, displaying mononuclear on the lower left and polymorphonuclear on the upper right. C, The separation and plotting of the
polymorphonuclear cells into neutrophils and eosinophils based on MAPPS technology. The granularity/lobularity is demonstrated by the X axis plotted at 90
degrees scatter and the Y axis at 90 degrees D scatter. The size and complexity of the cells are plotted on the X axis at 10 degrees and the Y axis at 0
degrees. The system uses algorithms to further separate the two populations of cells. D, The scatter of all WBC populations by MAPPS technology looking at
the cells by granularity/lobularity and size and complexity. 90 D, 90 depolarized. (From Abbott Laboratories: CELL-DYN 3700 system operators manual
[914032C], Abbott Park, Ill, 2000.)

hemoglobin concentration (x-axis) cytogram or RBC map platelets from interfering particles, such as RBC fragments and
(Figure 39-11).20 small RBCs. Larger platelets can be included in the platelet
Independent histograms of RBC volume and hemoglobin count.24,48
concentration also are plotted. On the Technicon H systems, Several parameters and indices are derived from the mea-
platelets are counted and sized from the high-angle signals only, surements described in the previous paragraph. MCV and MPV
and a platelet size-distribution histogram is generated.23 The are the mean of the RBC volume histogram and the platelet
ADVIA 2120 and 120 use two-dimensional (low-angle and high- histogram. Hematocrit, MCH, and MCHC are mathematically
angle) platelet analysis, which allows better discrimination of computed using RBC, hemoglobin, and MCV values. RDW is
610 PART VII Cell-Counting Automation

CELL-DYN 3700

A B
G
R
A
S N
WBC 8.80 K/uL I U
NEU 5.58 63.4 %N Z L
LYM 2.14 24.4 %L
MONO .970 11.0 %M
E A
EOS .058 .662 %E R
BASO .045 .516 %B I
T
Y
RBC 4.18 M/uL
HGB 12.1 g/dL COMPLEXITY LOBULARITY
HCT 36.7 %
MCV 87.9 fL
MCH 29.0 pg
MCHC 33.0 g/dL
D C
RDW 16.7 %

PLT 312. K/uL

RBC PLT

Figure 39-9 Composite scatterplots/histograms obtained from the CELL-DYN 3700 for the same normal sample analyzed in Figures 39-6, 39-7, and
39-12. A, White blood cell (WBC) data derived by multiangle polarized scatter separation (MAPSS) technology. Forward light scatter at 0 degrees, which
correlates with volume (y-axis) is plotted against 10-degree narrow-angle light scatter, which correlates with complexity (x-axis). Neutrophil (NEU), monocyte
(MONO), and lymphocyte (LYM) population clusters are represented. Not shown: 90-degree/10-degree scatterplot that separates the leukocytes into mono-
nuclear and segmented (polymorphonuclear) cell populations and 0-degree/10-degree scatterplot that separates the mononuclear population into lymphocytes,
monocytes, and basophils (BASO). (Basophils cluster with mononuclear cells because the cytoplasmic granules dissolve in the sheath fluid.) B, Separation of
the segmented population by plotting 90-degree depolarized light scatter (y-axis), which correlates with granularity, against 90-degree orthogonal light scatter
(x-axis), which correlates with lobularity or internal complexity. Eosinophils (EOS), because of their unique granulation, scatter more 90-degree depolarized
light. A dynamic threshold is used to determine the best separation between the eosinophil and neutrophil populations. Computer algorithms determine the
relative number (percentage) of each subpopulation. The absolute number (K/L) is calculated by multiplying the percentage of each subpopulation by the
WBC count. C and D, Platelet (PLT) and red blood cell (RBC) data determined in the impedance channel. The small population of larger RBCs on the right of
the RBC curve represents coincident passage of doublets or triplets or both through the aperture, which creates larger pulses. The red cell distribution width
(RDW) is derived from the 20th and 80th percentiles of the RBC histogram, so that these artificially large pulses are excluded from the calculation.

calculated as the CV of the RBC volume histogram, whereas not reported as a patient result but is used by the instrument
hemoglobin distribution width (HDW), an analogous index, as an internal check for the MCHC and is available to the
is calculated as the SD of the RBC hemoglobin concentration operator for calculating the cellular hemoglobin if interfer-
histogram. The reference interval for HDW is 2.2 to 3.2g/dL. ences are present. Unique RBC flags derived from CHCM
Cell hemoglobin concentration mean (CHCM), analogous to include hemoglobin concentration variance (HC VAR), hypo-
MCHC, is derived from cell-by-cell direct measures of hemo- chromia (HYPO), and hyperchromia (HYPER).23,24
globin concentration. Interferences with the hemoglobin colo- Siemens hematology analyzers determine WBC count and
rimetric method, such as lipemia or icterus, affect the calculated a six-part WBC differential (neutrophils, lymphocytes, mono-
MCHC but do not alter measured CHCM. CHCM generally is cytes, eosinophils, basophils, and large unstained cells [LUCs])
CHAPTER 39 Automated Cell-Counting Instrumentation 611

High-angle detector

Cell

Laser 0 angle

Low-angle
detector

A
Figure 39-10 Differential scatter detection as used in the Technicon H
systems and ADVIA 120. Forward high-angle scatter (5 to 15 degrees) and
forward low-angle scatter (2 to 3 degrees) are detected for analysis of red
and white blood cells. (From Miles: Technicon H systems training guide,
Tarrytown, NY, 1993, Miles.)

by cytochemistry and optical flow cytometry, using the PEROX


and BASO channels. LUCs include reactive or variant lympho-
cytes and blasts.

Peroxidase (PEROX) Channel B


In the PEROX channel, RBCs are lysed, and WBCs are stained Figure 39-11 Cytograms or red blood cell (RBC) maps derived using the
for their peroxidase activity. The following reaction is catalyzed Mie theory of light scatter of dielectric spheres. A, Transformation between
by cellular peroxidase, which converts the substrate to a dark scatter angles (2 to 3 degrees and 5 to 15 degrees) and RBC volume (V) and
precipitate in peroxidase-containing cells (neutrophils, mono- hemoglobin concentration (HC). B, RBC map for a patient sample. (From
cytes, and eosinophils): Groner W: New developments in flow cytochemistry technology. In Simson
E, editor: Proceedings of Technicon H-1 hematology symposium, October
11, 1985, Tarrytown, NY, 1986, Technicon Instruments Corp, p 5.)
H2O2 + 4-chloro-1-naphthol cellular
peroxidase
dark precipitate

A portion of the cell suspension is fed to a sheath-stream (including variant or reactive lymphs and blasts) contain no
flow cell where a tungsten-halogen darkfield optics system is peroxidase and appear on the left of the cytogram, with LUCs
used to measure absorbance (proportional to the amount of appearing above the lymphocyte area. Basophils cluster with
peroxidase in each cell) and forward scatter (proportional to the small lymphocytes and require further analysis for
the size of each cell). Absorbance is plotted on the x-axis of the classification.23,24,49,51
cytogram, and scatter is plotted on the y-axis.23,24 A total WBC
count (WBC-PEROX) is obtained from the optical signals in Basophil-Lobularity (BASO) Channel
this channel and is used as an internal check of the primary In the BASO channel, cells are treated with a reagent containing
WBC count obtained in the basophil-lobularity channel (WBC- a nonionic surfactant in an acidic solution. Basophils are par-
BASO). If significant interference occurs in the WBC-BASO ticularly resistant to lysis in this temperature-controlled reac-
count, the instrument substitutes the WBC-PEROX value.24 tion, whereas RBCs and platelets lyse and other leukocytes
Computerized cluster analysis allows classification of the (nonbasophils) are stripped of their cytoplasm. Laser optics,
different cell populations, including abnormal clusters such as using the same two-angle (2 to 3 degrees and 5 to 15 degrees)
nucleated RBCs and platelet clumps. Nucleated RBCs are forward scattering system of the RBC/platelet channel, is used
analyzed for every sample using four counting algorithms, to analyze the treated cells. High-angle scatter (proportional to
which permits the system to choose the most accurate count nuclear complexity) is plotted on the x-axis, and low-angle
based on internal rules and conditions. Neutrophils and eosin- scatter (proportional to cell size) is plotted on the y-axis.
ophils contain the most peroxidase and cluster to the right on Cluster analysis allows for identification and quantification of
the cytogram. Monocytes stain weakly and cluster in the mid- the individual cellular populations. The intact basophils are
region of the cytogram. Lymphocytes, basophils, and LUCs identifiable by their large low-angle scatter. The remaining
612 PART VII Cell-Counting Automation

nuclei are classified as mononuclear, segmented, and blast cell to stain residual RNA in the reticulocytes.* Because neither
nuclei based on their nuclear complexity (shape and cell the FACS nor EPICS system is generally available in the routine
density) and high-angle scatter.23,24,49 hematology laboratory, the discussion here is limited to the
Basophils fall above a horizontal threshold on the cyto- other analyzers.
gram. The stripped nuclei fall below the basophils, with seg- The Sysmex R-3000/3500 is a stand-alone reticulocyte ana-
mented cells to the right and mononuclear cells to the left lyzer that uses auramine O, a supravital fluorescent dye, and
along the x-axis. Blast cells uniquely cluster below the mono- measures forward scatter and side fluorescence as the cells
nuclear cells. Lack of distinct separation between the segmented pass through a sheath-stream flow cell past an argon laser.
and mononuclear clusters indicates WBC immaturity or sus- The signals are plotted on a scattergram with forward scatter
pected left shift. As indicated earlier, this channel provides intensity, which that correlates with size, plotted against
the primary WBC count, the WBC-BASO. Relative differential fluorescence intensity, which is proportional to RNA content.
results (in percent) are computed by dividing absolute numbers Automatic discrimination separates the populations into
of the different cell classifications by the total WBC count.23,24,49 mature RBCs and reticulocytes. The reticulocytes fall into
The nucleated RBC method is based on the physical char- low-fluorescence, middle-fluorescence, or high-fluorescence
acteristics of size and density of the nucleated RBC nuclei. regions, with the less mature reticulocytes showing higher fluo-
These characteristics allow counting in both WBC channels rescence. The immature reticulocyte fraction (IRF) is the sum of
on the ADVIA 2120, and algorithms are applied to determine the middle-fluorescence and high-fluorescence ratios and indi-
the absolute number and percentage of nucleated RBCs. Infor- cates the ratio of immature reticulocytes to total reticulocytes
mation from the PEROX and BASO channels is used to in a sample. The XE-5000 has a parameter called RET-He that
generate differential morphology flags indicating the possible measures the hemoglobin content of the reticulocytes; this is
presence of atypical (reactive) lymphocytes, blasts, left shift, similar to the reticulocyte hemoglobin content (CHr) param-
immature granulocytes, nucleated RBCs, or large platelets or eter on the ADVIA 2120i.37,39,60 Platelets, which also are
platelet clumps.23,24,51 Figure 39-12 shows a patient printout counted, fall below a lower discriminator line.57 The Sysmex
from the ADVIA 120. SE-9500/9000+RAM-1 module uses the same flow cytometry
methodology for reticulocyte counting as the R-3500.16 Off-
line sample preparation is not required. The smaller Sysmex
AUTOMATED RETICULOCYTE COUNTING
R-500 uses flow cytometry with a semiconductor laser as the
Reticulocyte counting is the last of the manual cell-counting light source and polymethine supravital fluorescent dye to
procedures to be automated and has been a primary focus of provide automated reticulocyte counts.29
hematology analyzer advancement in recent years. The impre- The CELL-DYN 3500R performs reticulocyte analysis by
cision and inaccuracy in manual reticulocyte counting is due measuring 10-degree and 90-degree scatter in the optical
to multiple factors, including stain variability, slide distribu- channel (MAPSS technology) after the cells have been isovolu-
tion error, statistical sampling error, and interobserver error.52 metrically sphered to eliminate optical orientation noise. The
All of these potential errors, with the possible exception of RBCs are stained with the thiazine dye new methylene blue N
stain variability, are correctable with automated reticulocyte in an off-line sample preparation before the sample is intro-
counting. Increasing the number of RBCs counted produces duced to the instrument. The operator simply must change
increased precision.53 This was evidenced in the 1993 College computer functions on the instrument before aspiration of the
of American Pathologists pilot reticulocyte proficiency survey reticulocyte preparation.46 The CELL-DYN Sapphire also uses
(Set RT-A, Sample RT-01) on which the CV for the reported MAPSS technology but adds fluorescence detection to allow
manual results was 35% compared with 8.3% for results fully automated, random access reticulocyte testing.25,47,41 The
obtained using flow cytometry.54 Precision of automated RBCs are stained with a proprietary membrane-permeable fluo-
methods has continued to improve. The manual reticulocyte rescent dye (CD4K530) that binds stoichiometrically to nucleic
results for one sample in the 2000 Reticulocyte Survey Set RT/ acid and emits green light as the cells pass through a sheath-
RT2-A showed a CV of 28.7%, whereas the CV was 2.8% for stream flow cell past an argon ion laser. Platelets and reticulo-
results obtained using one of the automated methods.55 Auto- cytes are separated based on intensity of green fluorescence
mated reticulocyte analyzers may count 32,000 RBCs com- (scatter measured at 7 degrees and 90 degrees), and the reticu-
pared with 1000 cells in the routine manual procedure.56 locyte count along with the IRF is determined.47,61
Available automated reticulocyte analyzers include flow Beckman Coulter also has incorporated reticulocyte
cytometry systems such as the FACS system from Becton, methods into its primary cell-counting instrumentsthe STKS
Dickinson and Company (Franklin Lakes, N.J.) or the Coulter and the MAXM and LH 700 series systems. Off-line staining
EPICS system; the Sysmex R-3500, R-500, XE-2100, and preparations are required for the STKS and MAXM, but the
XE-5000 systems; the CELL-DYN 3500R, 3700, and 4000 same laser optics employed in cell counting are used for reticu-
systems; the Coulter GEN-S, STKS, MAXM, and LH 750 systems; locyte enumeration. The Coulter method uses a new methylene
and the Siemens ADVIA 2120, 2120i, and 120 and Technicon blue stain and the VCS technology described earlier. Volume
H-3 RTC and RTX systems. All of these analyzers evaluate is plotted against light scatter (DF 5 scatterplot) and against
reticulocytes based on optical scatter or fluorescence after the
RBCs are treated with fluorescent dyes or nucleic acid stains *References 16, 23, 24, 35, 37, 46, 47, 56-59.
CHAPTER 39 Automated Cell-Counting Instrumentation 613

Header - WB Morphology Flags A Perox B Baso


SID: 4004 MICRO:
Time & Date: 5/25/05 10:20:38AM MACRO:
HYPO:
Selectivity: CBC\DIFF\RETIC HYPER:
Sample Type: PATIENT ANISO:
Sex & Age: HC VAR:
System: 30810242 LEFT SHIFT:
Sequence: 613 BLASTS:
Species: Human ATYPS:
NRBC:
IG:
FOR LABORATORY USE ONLY LARGE PLT:
PLT CLUMPS:
MPO DEF:
RBC FRAG:
Routine CBC RBC GHOSTS:

WBC: 8.36 x103 cells/L


RBC: 4.30 x106 cells/L
HGB: 12.5 g/dL RBC HC C RBC V/HC
HCT: 38.0 %
MCV: 88.4 fL
MCH: 29.1 pg
MCHC: 32.9 g/dL
CHCM: 32.7 g/dL
RBC Volume
CH: 28.7 pg
RDW: 14.2 %
HDW: 2.32 g/dL
PLT: 332 x103 cells/L
MPV: 7.4 fL
Platelet Vol
Routine WBC Differential
% x103 cells/L
WBC: 8.36
Neut: 69.3 5.79
Lymph: 21.7 1.81
Mono: 6.5 0.55
EOS: 0.9 0.08
Baso: 0.3 0.03
LUC: 1.2 0.10
LI: 1.96
MPXI: -4.3
WBCP: 8.88

Figure 39-12 Composite scatterplots/histograms obtained from the ADVIA 120 for the same normal specimen analyzed in Figures 39-6, 39-7, and 39-9.
A, White blood cell (WBC) data derived from the peroxidase (Perox) channel in which the WBCs have been stained for their peroxidase activity. Darkfield optical
scatter, which is correlated with volume, is plotted on the y-axis against light absorbance, which is proportional to staining intensity, on the x-axis. Computer
cluster analysis separates the WBCs into neutrophils (Neut), eosinophils (EOS), monocytes (Mono), lymphocytes (Lymph), and large, unstained cells (LUC) as
indicated. Basophils cluster with small lymphocytes in the Perox channel. B, WBC data derived from the laser-based basophil-lobularity (Baso) channel in
which the cells have been treated with a differential lysis reagent. Low-angle (2- to 3-degree) scatter, which correlates with size, is plotted on the y-axis
against high-angle (5- to 15-degree) scatter, which correlates with nuclear complexity (shape and cell density), on the x-axis. Nonlysed basophils, which have
large low-angle scatter, fall above the stripped nuclei (nonbasophils), which cluster into segmented (polymorphonuclear) and mononuclear populations. Blasts,
when present, uniquely cluster below the mononuclear population. Basophils are subtracted from the lymphocyte count for the final absolute counts ( 103/L).
Relative differential results (percentages) are computed by dividing absolute counts by the total WBC count. C, RBCs also are analyzed using the Mie theory
of light scatter. This RBC volume/hemoglobin (Hb) concentration cytogram represents a linear plot of the RBC analysis. Volume (V) is plotted on the y-axis
against Hb concentration (HC) on the x-axis. The direct measure of Hb concentration allows classification of RBCs as macrocytic, normocytic, or microcytic
and as hypochromic, normochromic, or hyperchromic. Single-parameter histograms for RBC Hb concentration, RBC volume, and platelet volume are generated.
The RBC histogram has markers at the 60fL and 120fL channels for visualization of microcytosis or macrocytosis or both. Likewise, the RBC HC curve has
markers at the 28g/dL and 41g/dL channels for visualization of hypochromia and hyperchromia. Hb distribution width, analogous to the RDW, is derived
from the Hb concentration histogram.
614 PART VII Cell-Counting Automation

conductivity (DF 6 scatterplot), which correlates with opacity medium-absorbing and high-absorbing cells reflects the IRF.
of the RBC. Stained reticulocytes show greater optical scatter Volume and hemoglobin concentration for each cell are
and greater opacity than mature RBCs. Relative and absolute derived from the RBC map by applying Mie scattering
reticulocyte counts are reported, along with mean reticulocyte theory.26,62 Unique reticulocyte indices (MCVr, CHCMr, RDWr,
volume and maturation index or IRF.56 HDWr, CHr, and CHDWr) are provided. The CHr of each cell
The Siemens ADVIA 2120, 2120i, and 120 and Technicon is calculated as the product of the cell volume and the cell
H-3 RTC or RTX systems enumerate reticulocytes in the same hemoglobin concentration. A single-parameter histogram of
laser optics flow cell used in the RBC/platelet and BASO chan- CHr is constructed, with a corresponding distribution width
nels described earlier. The H-3 systems require an off-line (CHDWr) calculated.23,24 These reticulocyte indices are not
sample preparation. The reticulocyte reagent isovolumetrically reported on the routine patient printout but are available to
spheres the RBCs and stains the reticulocytes with oxazine 750, the operator. Figure 39-13 is a reticulocyte printout from an
a nucleic acidbinding dye. Three detectors measure low-angle ADVIA 120, showing the cytograms and reticulocyte indices.
scatter (2 to 3 degrees), high-angle scatter (5 to 15 degrees), Automation of reticulocyte counting has allowed increased
and absorbance simultaneously as the cells pass through the precision and accuracy and has greatly expanded the analysis
flow cell. Three cytograms are generated: (1) high-angle scatter of immature RBCs, providing new parameters and indices that
versus absorption, (2) low-angle scatter versus high-angle may be useful in the diagnosis and treatment of anemias. The
scatter (Mie cytogram or RBC map), and (3) volume versus IRF is a reliable indicator of changes in erythropoietic activity
hemoglobin concentration. The absorption cytogram allows and may prove to be a valuable therapeutic monitoring tool
separation and quantitation of reticulocytes, with additional in patients with chronic renal failure.63,64 The reticulocyte
subdivision into low-absorbing, medium-absorbing, and high- maturity measurements also may be useful in evaluating
absorbing cells based on amount of staining. The sum of the bone marrow suppression during chemotherapy, monitoring

ADVIA 120

I
Retic A Retic Scatter Abs B Retic Scatter

% # Coincidence events
Neg RBC: 98.9 37406
Retic: 1.1 46.6
L Retic: 88.6 366
M Retic: 10.2 42
H Retic: 1.2 5
IRF-H: 1.2
IRF-M+H: 11.4 Med abs retics
RTC Cells Acquired: 41294
RTC Cells Analyzed: 39076
RTC Gated Cells: 37819
Retic Count: 413 Mature RCBs High abs retics
Low abs retics
Platelets

II
Retic C Retic Scatter Abs D Retic Scatter

% #
Neg RBC: 87.6 32015
Retic: H 12.4* H 519.2*
L Retic: 72.3 3278
M Retic: 21.4 968
H Retic: 6.3 286
IRF-H: 6.3
IRF-M+H: 27.7
RTC Cells Acquired: 42910
RTC Cells Analyzed: 37486
RTC Gated Cells: 36547
Retic Count: 4532

Figure 39-13 Composite cytograms obtained from the ADVIA 120. I, Normal reticulocyte count. II, High reticulocyte count with a large immature reticulocyte
fraction (IRF). I-A and II-C, Reticulocyte scatter/absorption cytograms show high-angle (5- to 15-degree) scatter on the y-axis versus absorption on the x-axis,
which allows separation of low-absorbing, medium-absorbing, and high-absorbing cells based on the amount of staining with nucleic acidbinding dye (oxazine
750). I-B and II-D, Reticulocyte scatter cytograms show low-angle (2- to 3-degree) scatter on the y-axis versus high-angle (5- to 15-degree) scatter on the
x-axis (red blood cell [RBC] map). Because the RBCs are evaluated by the instrument on a cell-by-cell basis, unique reticulocyte indices can be derived.
CHAPTER 39 Automated Cell-Counting Instrumentation 615

hematopoietic regeneration after bone marrow or stem cell with reference methods or review of quality control data after
transplantation, monitoring renal transplant engraftment, and calibration and by external comparison studies such as profi-
assessing efficacy of anemia therapy.63-69 The additional reticu- ciency testing.1,78
locyte indices derived on the ADVIA 2120 and 120 are valuable
in following the response to erythropoietin therapy, and the Instrument Limitations
CHr in particular has proved useful in the early detection and The continual improvement of automated technologies has
diagnosis of iron deficient erythropoiesis in children.65,69,70 resulted in greater sensitivity and specificity of instrument flag-
Widespread use of the new parameters may be limited by the ging with detection of possible interferences in the data. The
availability of instrumentation. parallel improvement in instrument walk-away capabilities has
increased the importance of the technologists awareness and
understanding of instrument limitations, however, and of his
LIMITATIONS AND INTERFERENCES
or her ability to recognize factors that may interfere and cause
Implementing automation in the hematology laboratory erroneous laboratory results. Limitations and interferences
requires critical evaluation of the instruments methods and may be related to methodology or to inherent problems in the
limitations, and performance goals for the individual labora- blood sample.
tory. The Clinical and Laboratory Standards Institute (CLSI; Each instrument has limitations related to methodology
formerly National Committee for Clinical Laboratory Stan- that are defined in instrument operation manuals and in the
dards) has approved a standard for performance goals for the literature. A common method limitation is an instruments
internal quality control of multichannel hematology analyz- inability to distinguish cells reliably from other particles or cell
ers.71 This standard provides guidelines for instrument calibra- fragments of the same size. Cell fragments may be counted as
tion and assessment of performance criteria, including accuracy, platelets in samples from chemotherapy-treated patients with
precision, linearity, sensitivity, and specificity. The clinical increased WBC fragility.1 Likewise, schistocytes or small RBCs
accuracy (sensitivity and specificity) of the methods should be may interfere with the platelet count. Larger platelet clumps
such that the instrument appropriately identifies patients who may be counted as WBCs, which results in a falsely decreased
have disease and patients who do not have disease.72 Quality platelet count and potentially increases the WBC count. Micro-
control systems should reflect the laboratorys established per- megakaryocytes may be counted as nucleated RBCs or WBCs.
formance goals and provide a high level of assurance that the RBCs containing variant hemoglobins such as Hb S or Hb C
instrument is working within its specified limits. are often resistant to lysis, and the unlysed cells can be falsely
counted as nucleated RBCs or WBCs and interfere in the hemo-
Calibration globin reaction.79 This phenomenon has become more appar-
Calibration is crucial in defining the accuracy (as opposed to ent with the use of gentler diluent and lysing reagents in the
precision; see Chapter 5) of the data produced. Calibration, or analyzers with automated WBC differential technology. Nonly-
the process of electronically correcting an instrument for ana- sis also may be seen in specimens from patients with severe
lytical bias (numerical difference from the true value), may liver disease, those undergoing chemotherapy treatment, and
be accomplished by appropriate use of reference methods, neonates (due to increased levels of Hb F) on the older Sysmex
reference materials, or commercially prepared calibrators.71 instruments15 and in samples from patients with markedly
Because few instruments are precalibrated by the manufacturer, elevated serum urea nitrogen levels on the Technicon H instru-
calibration must be performed at initial installation and veri- ments.23 The Technicon H systems are able to provide a correct
fied at least every 6 months under the requirements of the WBC count from the BASO channel (i.e., the WBC-BASO) with
Clinical Laboratory Improvement Act of 1988.73 Periodic reca- its stronger lysing reagent. The ADVIA 2120 and 120 reports
libration may be required after major instrument repair requir- the WBC-BASO as the primary WBC count.24 An extended lyse
ing optical alignment or part replacement. cycle may be used on the CELL-DYN 3500, and the newer
Whole-blood calibration using fresh whole-blood speci- instruments are able to provide a correct WBC impedance
mens requires the use of reference methods, materials, and count when lyse-resistant RBCs are present.22,57 The Sysmex
procedures to determine true values.1,74,75 The International SE-9000 and Sysmex SE-9500 also have an additional WBC
Committee for Standardization in Haematology has estab- impedance channel.16,80
lished guidelines for selecting a reference blood cell counter Suppression of automated data, particularly WBC differen-
for this purpose,1 but the cyanmethemoglobin method remains tial data, may occur when internal instrument checks fail or
the only standard available in hematology for calibration and cast doubt on the validity of the data. Instruments from some
quality control.76 Whole-blood calibration, which historically manufacturers release results with specific error codes or flag-
has been considered the preferred method for calibration of ging for further review. The rejection rate of the different instru-
multichannel hematology analyzers, has been almost com- ments varies, with the Coulter and Sysmex instruments showing
pletely replaced by the use of commercial calibrators assayed the highest rate of data suppression (10.4% and 18%, respec-
using reference methods. Calibration bias is possible with the tively) and the Bayer (Technicon) and CELL-DYN analyzers the
use of these calibrators because of inherent differences in sta- lowest (1.2% and 0%, respectively) in one study.81 The sup-
bilized and preserved cell suspensions.77 It is essential that pression of automated differential data ensures that a manual
calibrations be carried out properly and verified by comparison differential count is performed, whereas the release of data
616 PART VII Cell-Counting Automation

with appropriate flagging mandates the need for careful review and lipemia. Cold agglutinins manifest as a classic pattern of
of the data and possibly a blood film examination. This sug- increased MCV (frequently greater than 130fL), markedly
gests a difference in philosophy among the manufacturers and decreased RBC count, and increased MCHC (frequently greater
affects the work flow in different ways.81 More importantly, than 40g/dL). Careful examination of the histograms or
each laboratory must establish its own criteria for directed cytograms from the instruments may yield clues to this
blood film review based on established performance goals, abnormality.83 Icterus and lipemia directly affect hemoglobin
instrument flagging, and inherent instrument limitations.82 measurements and related indices.79 Table 39-3 summarizes
conditions that cause interference on most hematology analyz-
Sample Limitations ers and offers suggestions for manually obtaining correct
Limitations resulting from inherent sample problems include patient results. As instrumentation advances, instrumentation
those related to the presence of cold agglutinins, icterus, software can adjust or correct for some of the conditions listed.

TABLE 39-3 Conditions That Cause Interference on Most Hematology Analyzers


Condition Parameters Affected* Rationale Instrument Indicators Corrective Action
Cold agglutinins RBC , MCV , MCHC Agglutination of RBCs Dual RBC population on RBC Warm sample to 37 C and
, grainy appearance map, or right shift on RBC rerun
histogram
Lipemia, icterus, Hb , MCH Turbidity affects Hb 3 Hct 3, abnormal Plasma replacement
chylomicrons spectrophotometric reading histogram/cytogram
Hemolysis RBC , Hct RBCs lysed and not counted Hb 3 Hct 3, may Request new sample
show lipemia pattern on
histogram/cytogram
Lysis-resistant RBCs WBC , Hb RBCs with Hb S, C, or F may Interference at noise-WBC Perform manual dilutions,
with abnormal Hbs fail to lyse and be counted interface on histogram/ allow incubation time for
as WBCs cytogram lysis
Microcytosis or RBC Size of RBCs or RBC Left shift on RBC histogram, Review film
schistocytes fragments < lower RBC MCV flagged if below
threshold limits
Nucleated RBCs, WBC Nucleated RBCs or Nucleated RBC flag resulting Count nucleated RBCs or
megakaryocyte micromegakaryoblasts from interference at micromegakaryoblasts per
fragments, or counted as WBCs noise-lymphocyte interface 100 WBCs and correct
micromegakaryoblasts on histogram/cytogram
Platelet clumps PLT , WBC Large clumps counted as Platelet clumps/N flag, Redraw specimen in sodium
WBCs interference at noise- citrate, multiply result
lymphocyte interface on by 1.1
histogram/cytogram
WBC > 100,000/mcL Hb , RBC , Hct Turbidity on Hb, WBCs Hb 3 Hct 3, WBC Spun Hct, perform manual Hb
incorrect, abnormal counted with RBC count count may be above (spin/read supernatant),
indices linearity correct RBC count,
recalculate indices; if above
linearity, dilute for correct
WBC count
Leukemia, especially Spurious WBC , Fragile WBCs, fragments Platelet count inconsistent Review film, perform phase
with chemotherapy spurious PLT counted as platelets with previous results platelet count
Old specimen MCV , MPV , PLT , RBCs swell as sample ages, Abnormal clustering on WBC Establish stability and sample
automated differential platelets swell and histogram/cytogram rejection criteria
may be incorrect degenerate, WBCs affected
by prolonged exposure to
EDTA

, Increased; , decreased; EDTA, ethylenediamine tetraacetic acid; Hb, hemoglobin; Hct, hematocrit; MCH, mean cell hemoglobin; MCHC, mean cell hemoglobin concentration;
MCV, mean cell volume; MPV, mean platelet volume; PLT, platelet count; RBC, red blood cell (or count); WBC, white blood cell (or count).
*Manufacturers labeling.

Lipemia shows signature pattern on Siemens ADVIA 120 and Technicon H cytograms.

Hb can be back-calculated from directly measured MCHC on Siemens ADVIA 120 and Technicon H cytograms.

Small nucleated RBCs thresholded out of WBC count on Sysmex SE-9000 and CELL-DYN 3500; correction for nucleated RBCs may not be necessary. CELL-DYN 4000 and
Sysmex XE-2100 directly measure nucleated RBCs, which are gated out of the WBC count.29,47 Semiautomated Sysmex instruments have adjustable lower thresholds to allow
inclusion of all nucleated cells in the WBC count.127
CHAPTER 39 Automated Cell-Counting Instrumentation 617

Historically, a nucleated RBC flag required examination of a in populations with abnormalities.93-95 The five-part automated
blood film to enumerate the nucleated RBCs and correct the differentials available on the larger instruments have been
WBC. All four major vendors offer online nucleated RBC evaluated extensively and have acceptable clinical sensitivity
enumeration and WBC correction, although the laboratory and specificity for detection of distributional and morphologic
must validate the results. Lipemia interferes with the hemo abnormalities.43,44,96-103 Abnormal cells such as blasts and
globin reading by falsely elevating the hemoglobin and nucleated RBCs in low concentrations may not be detected by
associated indices. The Siemens technology uses direct mea- the instruments but likewise may be missed by the routine
surement of the CHCM parameter, which allows back- 100-cell manual/visual differential count.104,105 The CELL-DYN
calculation of the hemoglobin unaffected by lipemia and thus Sapphire, with its added fluorescent detection technology, has
eliminates the need for the manual method of saline replace- been shown to have high sensitivity and specificity for flagging
ment in lipemic samples. These two examples involving nucleated RBCs and platelet clumps.41,50,106 As technology con-
nucleated RBCs and lipemia illustrate instrument advances, tinues to improve, blood film review to confirm the presence
and continued future improvements in technology will elimi- of platelet clumps or nucleated RBCs and to correct leukocyte
nate or decrease the need for manual intervention to obtain counts for interference from platelet clumps or nucleated
accurate results. RBCs is becoming unnecessary, especially for nucleated RBCs,
Sample age and improper sample handling can have pro- because the four major vendors now count and correct the
found effects on the reliability of hematology test results. These WBC for nucleated RBCs on their high-end analyzers.*
factors have even greater significance as hospitals move toward Instrument evaluations based on the CLSI H20-T or H20-A
greater use of off-site testing by large reference laboratories. standard (Reference Leukocyte Differential Count [Propor-
Specific problems with older samples include increased WBC tional] and Evaluation of Instrumental Methods)107 using an
fragility, swelling and possible lysis of RBCs, and the deteriora- 800-cell or 400-cell manual leukocyte differential count as the
tion of platelets.15 Stability studies should be performed before reference method have shown acceptable correlation coeffi-
an instrument is used, and specific guidelines should be estab- cients for all WBC types, with the possible exception of mono-
lished for specimen handling and rejection. cytes.44,81,108-110 However, further studies using monoclonal
antibodies as the reference method for counting monocytes
suggest that automated analyzers yield a more accurate assess-
CLINICAL UTILITY OF AUTOMATED
ment of monocytosis than do manual methods.111,112
HEMATOLOGY INSTRUMENTATION
Histograms and cytograms along with instrument flagging
The use of automated cell-counting analyzers has directly provide valuable information in the diagnosis and treatment
affected the availability, accuracy, and clinical usefulness of the of RBC and WBC disorders. Multiple reports indicate the use-
CBC and WBC differential count. Some parameters that are fulness of histograms and cytograms in the characterization of
available on hematology instrumentation, but cannot be various abnormal conditions, including RBC disorders such as
derived manually, have provided further insight into various cold agglutination and WBC diseases such as leukemias and
clinical conditions. The RDW, a quantitative estimate of eryth- myelodysplastic disorders.83,113-115 The major instrument manu-
rocyte anisocytosis, has been used with the MCV in anemia facturers have published case study books with histograms and
classification. RDW has been shown to be unreliable as a sole cytograms to aid the technologist in the interpretation of
discriminator of microcytic anemias.84 The direct measurement instrument data.116-120
of RBC hemoglobin concentration on the Siemens systems has Manufacturers are developing integrated hematology work-
proved to be a valuable tool in the early detection of iron stations for the greatest automation and laboratory efficiency.
deficiency anemia and in the discrimination of iron deficiency The Sysmex Total Hematology Automation System (HST series)
from thalassemia minor.85,86 Directly measured hemoglobin robotically links the SE-9000, R-3500 (automated reticulocyte
concentration also has been used to detect iron-deficient eryth- analyzer), and SP-100 (automatic slide maker/stainer). The
ropoiesis in iron-replete subjects treated with recombinant HST line links two XE-2100 units and one SP-100 instrument
erythropoietin.87 The MPV may be useful in detecting whether for complete automation or systemization of hematology
or not the bone marrow is working correctly. A low MPV may testing. The SE-Alpha is a smaller version that links the SE-9000
indicate marrow hypoproduction, whereas a high MPV may and SP-100.29 The Siemens ADVIA LabCell links the ADVIA
raise suspicion of an abnormal myeloproliferative disorder.88 2120 to the track, and the Autoslide (automatic slide maker/
Methodology, anticoagulation, and storage time all influence stainer) links to the ADVIA 2120. The other manufacturers
the MPV, however, which makes the parameter less useful.89 have or are developing similar automation systems.121 Finally,
Automation of the WBC differential has had a significant as a result of increasing customer needs, manufacturers are
impact on the laboratory work flow because of the labor- adding body fluid counting to their high-end instrumentation.
intensive nature of the manual differential count. The three- The Beckman Coulter LH 780 and the Sysmex XE-5000 count
part differential available on earlier instruments generally WBCs and RBCs in body fluids, and the ADVIA 2120i/120
proved suitable as a screening leukocyte differential count to counts WBCs and RBCs in cerebrospinal fluid and performs a
identify samples that required further workup or a manual differential count on cerebrospinal fluid.37,122,123
differential count.90-92 It has been shown, however, that partial
differential counts do not substitute for a complete differential *References 34, 37, 41, 44, 61, 106.
618 PART VII Cell-Counting Automation

Selection of a hematology analyzer for an individual labora- slide makers/stainers can be programmed to make blood films
tory requires careful evaluation of the laboratorys needs and for every sample or to make films based on the laboratorys
close scrutiny of several important instrument issues, including internal criteria for a film review. This reduces, but does not
instrument specifications and system requirements, methods completely eliminate, the use of manual processes in the
used, training requirements, maintenance needs, reagent hematology laboratory. Automated slide makers/stainers do
usage, data management capabilities, staff response, and short- not connect to every analyzer and are not suitable for some
term and long-term expenditures.124 All instruments claim to laboratories. The instrument selected should suit the workload
improve laboratory efficiency through increased automation and patient population and should have a positive effect on
that results in improved work flow and faster turnaround time patient outcomes.125 The instrument selected for a cancer center
or through the addition of new parameters that may have clini- may be different from that chosen for a community hospital.126
cal efficacy. All four major vendors offer a slide maker/stainer Ultimately, however, the instrument decision may be swayed
that can be connected directly to their high-end analyzers. The by individual preferences.81

SUMMARY
Automated cell counting provides greater accuracy and greater Major manufacturers of hematology instrumentation include
precision than manual cell-counting methods. Beckman Coulter, Inc.; Sysmex Corporation; Abbott Diagnostics;
The primary principles of operation, electronic impedance and Siemens Healthcare Diagnostics, Inc. Beckman Coulter instru-
and optical scatter, are used by most automated hematology ments primarily use the impedance method of counting and sizing
analyzers. RF is sometimes used in conjunction with electronic cells; Sysmex instruments, a combination of impedance and RF;
impedance. Abbott instruments, impedance and optical measurements; and
The electronic impedance method detects and measures changes Siemens instruments, optical flow cytometry.
in electrical resistance between two electrodes as cells pass Reticulocyte analysis has been incorporated into the primary cell-
through a sensing aperture. The measurable voltage changes are counting instruments of all major manufacturers. All use either
plotted on frequency distribution graphs, or histograms, that allow fluorescent dyes or nucleic acid stains to stain reticulocytes before
the evaluation of cell populations based on size. the cells are counted using fluorescence or absorbance.
RF resistance uses high-voltage electromagnetic current. Measur- Each instrument has limitations related to methodology that may
able changes in the RF signal are proportional to cell interior result in instrument flagging of specific results or suppression of
density, or conductivity. Impedance and conductivity can be automated data. Likewise, inherent sample problems may result
plotted against each other on a two-dimensional distribution cyto- in instrument flagging that indicates possible rejection of auto-
gram or scatterplot, which allows the evaluation of cell populations mated results.
using cluster analysis. Automation of cell counting has had a significant impact on labora-
Optical scatter systems (flow cytometers) use detection of interfer- tory work flow, particularly automation of the WBC differential. In
ence in a laser beam or light source to differentiate and enumerate addition, newer parameters that can now be measured, such as
cell types. RDW, IRF, and CHr, have documented clinical utility.

R E V I E W Q UESTIONS
Examine the histograms/scatterplots obtained from four major c. Coulter
instruments for the same patient sample (Figure 39-14). d. Sysmex
Compare the results and respond to questions 1 to 4 based on
the results. 3. What do you suspect is the cause of the variation in platelet
flagging?
1. The manual differential showed the presence of nucleated a. Some instruments have higher levels of sensitivity.
RBCs and blasts. Which instrument flagged for the possi- b. All instruments use the same principle for counting
ble presence of blasts? platelets.
a. ADVIA c. Some instruments are susceptible to false-positive
b. CELL-DYN platelet flagging under certain conditions.
c. Coulter d. Some instruments have higher values for the lower
d. Sysmex limit of the reference range.

2. Which instrument did not alert the operator to the possi- 4. Based on the overall flagging for this sample on each
bility of nucleated RBCs? instrument, should a manual differential count be per-
a. ADVIA formed for this patient? Explain.
b. CELL-DYN
CHAPTER 39 Automated Cell-Counting Instrumentation 619

Sample ID: 2002 Cass / Pos: 000902 Operator ID: LABADMIN


Date: 5/26/2005
Time: 12:28:55 Sample Type: CD A No Read Listname: 37H5QDE2 Instrument: LHIN

Suspect Definitive
WBC Histogram Cellular Interference
WBC 5.3 R 3
10 /L
NE % %

LY % %

MO % % 50 100 200 300 PL

EO % %

BA % %

NRBC % %

NE #

103/L

LY #

103/L

MO # 103/L

103/L
EO #
103/L
BA #

NRBC # 103/L

RBC Histogram
RBC 3.03 L 106/L
HGB 9.4 L g/dL

HCT 26.9 L %

MCV 88.8 fL 50 100 200 300 PL

MCH 31.0 pg

MCHC 34.9 g/dL

RDW 14.9 H %

PLT Histogram
PLT 23 RCL 103/L
MPV 11.0 R H fL

2 10 20 30 PL

RET % 4.25
RET #
IRF 0.61

A Comments: UMBC = 5.9 [REVIEW SMEAR] [CALL RESULT]

Figure 39-14 Composite scatterplots/histograms obtained from four major instruments. A, Coulter LH 750.
Continued
620 PART VII Cell-Counting Automation

RBC V/HC Perox


Header-WB
SID: 040002265
Time & Date: 5/25/05 7:42:46 AM
Rack & Position: 3 1
Selectivity: CBC\DIFF
Sample Type: PATIENT
Sex & Age:
System: 30740242
Sequence: 373
Species: Human

FOR LABORATORY USE ONLY

Routine CBC
WBC: 6.31 * X103 cells/uL
RBC: L 3.14 X106 cells/uL
HGB: L 9.3 g/dL
HCT: L 27.2 %
MCV: 86.8 fL Morphology Flags Baso
MCH: 29.6 pg
MICRO:
MCHC: 34.1 g/dL
MACRO:
CHCM: 35.8 g/dL
YHYPO:
CH: 30.7 pg
HYPER:
RDW: H 17.5 % RBC HC ANISO:
HDW: H 3.68 g/dL
HC VAR:
PLT: L 21 X103 cells/uL +
LEFT SHIFT:
MPV: 8.1 fL +
BLASTS:
ATYPS: +
Routine WBC Differential NRBC: ++ +
% X103 cells/uL IG
Platelet Vol LARGE PLT:
WBC: 6.31 *
PLT CLUMPS:
Neut: 54.7 * 3.45 *
MPO DEF:
Lymph: 28.7 * 1.81 * RBC FRAG:
Mono: H 10.5 * 0.66 * RBC GHOSTS:
Eos: 0.2 * 0.01 *
Baso: 0.8 0.05
LUC: H 5.1 * 0.32 *
LI: 2.35 *
MPXI: -9.3
WBCP: 5.42

Figure 39-14, contd B, ADVIA 120.

5. A patient film demonstrates agglutinated RBCs , and the c. Involves detection and measurement of changes in
CBC shows an elevated MCHC. What other parameters electrical current between two electrodes
will be affected by the agglutination of the RBCs? d. Uses the variation in electrical charges applied to cells
a. MCV will be decreased and the RBC count will be based on their surface markers
increased.
b. MCV will be decreased and the RBC count will be ______6. Impedance
decreased.
c. MCV will be increased and the RBC count will be ______7. RF
decreased.
d. MCV will be decreased and the RBC count will be ______8. Optical scatter
increased.
9. Low-voltage DC is used to measure:
For each of the cell-counting methods listed in 6 to 8 select a. Cell nuclear volume
the appropriate definition in a to d. b. Total cell volume
a. Uses diffraction, reflection, and refraction of light c. Cellular complexity in the nucleus
waves d. Cellular complexity in the cytoplasm
b. Uses high-voltage electrical waves to measure the
internal complexity of cells
CHAPTER 39 Automated Cell-Counting Instrumentation 621

Positive
Diff. Morph.
Count
DIFF WBC/BASO

FSC
SFL
WBC 5.94 * [103/uL]
RBC 2.98 - [106/uL]
HGB 9.1 - [g/dL]
HCT 26.9 - [%]
MCV 90.3 - [fL]
MCH 30.5 [pg]
MCHC 33.8 [g/dL] SSC SSC
PLT & 24 * [103/uL]
RDW-SD 46.5 [fL]
RDW-CV 15.2 [%]
RET PLT-O

FSC

FSC
MPV ---- [fL]
NEUT 2.77 * [103/uL] 46.7 * [%]
LYMPH 1.64 * [103/uL] 27.6 * [%]
MONO 1.45 * [103/uL] 24.4 * [%]
EO 0.00 * [103/uL] 0.0 * [%]
BASO 0.08 * [103/uL] 1.3 * [%]
RET 3.06 + [%] 0.0912 + [103/uL]
IRF 51.7 + [%]

SFL SFL

RBC PLT

RBC/RET IP Message(s) PLT IP Message(s)


WBC IP Message(s)
PLT Abn Distribution
Monocytosis
Thrombocytopenia

Immature Gran?
PLT Clumps?
Left Shift?
NRBC?
C

Figure 39-14, contd C, Sysmex XE-2100.

10. Orthogonal light scatter is used to measure: Match each instrument listed in a to d with the technology
a. Cell volume or size it uses to determine WBC differentials as shown in 12 to 15.
b. Internal complexity of the cell a. Abbott CELL-DYN
c. Cellular granularity b. ADVIA
d. Nuclear density c. Sysmex
d. Coulter LH 750
11. On the Coulter instruments, hematocrit is a calculated
value. Which of the following directly measured parame- ______12. VCS technology
ters is used in the calculation of this value?
a. RDW ______13. MAPSS technology
b. Hemoglobin
c. MCV ______14. Peroxidase, optical scatter, and absorbance
d. MCHC
______15. RF/DC detection
622 PART VII Cell-Counting Automation

Specimen ID 2002 May 25 2005 16:17


Patient Operator ID DH
Sex DOB Sequence # 4139
Dr Open Sampler
Param: 1 Limits: 1

G
SUSPECT R
A
WBC 4.95 K/uL ( WOC ) S N
NEU 3.10 62.6 %N IG/BAN I U
LYM .692 14.0 %L Z L
MONO 1.10 22.1 %M NRBC E A
EOS .001 .023 %E R
BASO .062 1.26 %B I
T
Y

RBC 3.03 M/uL


HGB 9.00 g/dL COMPLEXITY LOBULARITY
HCT 27.4 %
MCV 90.4 fL
MCH 29.7 pg
MCHC 32.9 g/dL
RDW 16.1 %

URI
PLT 27.6 K/uL

RBC PLT
PATIENT LIMITS SET 1
WBC 4.50-10.8 RBC 4.00-5.60 PLT 137.-400. MANUAL DIFFERENTIAL RBC MORPHOLOGY
NEU .800-5.00 37.2-77.2 %N HGB 12.0-18.0 MPV 6.40-10.1
LYM 1.30-3.90 14.6-46.9 %L HCT 36.0-54.8 NEU META NORMAL MICRO
MONO .200-.800 3.50-12.9 %N MCV 80.5-99.9
EOS 0.00-.800 .700-5.90 %E MCH 26.9-34.3 BAND MYELO PLYCHROM MACRO
BASO 0.00-.100 .100-1.90 %B MCKC 32.5-35.2
RDW12.6-16.9 LYMPH PRO HYPCHROM ANISO
INTERPRETATION
RBC RBC PLT MONO BLAST POIK BASOSTIP

SUSPECTED ABNORMAL POPULATIONS: EOSIN VAR LYM TARGET


Immature Granulocytes/Bands Nucleated RBC PLT Upper Region Interference
WIC WOC Delta Alert BASO TOXGRAN SPHERO NRBC
USER-DEFINED ABNORMALITIES: COMMENT:
Lymphopenia Anemia Thrombocytopenia
DIFF BY DATE
Monocytosis
D

Figure 39-14, contd D, CELL-DYN 4000.

REFERENCES
1. Koepke JA: Quantitative blood cell counting. In Koepke JA, Philadelphia, 1998, Lippincott-Raven Publishers, pp 519-
editor: Practical laboratory hematology, New York, 1991, 551.
Churchill Livingstone, pp 43-60. 4. Coulter WH: High speed automatic blood cell counter
2. Rappaport ES, Helbert B, Beissner RS: Automated hematol- and cell size analyzer. Proc Natl Electron Conf 12:1034,
ogy: where we stand. South Med J 81:365-370, 1988. 1956.
3. Dotson MA: Multiparameter hematology instruments. In 5. Coulter Electronics: Significant advances in hematology: hema-
Stiene-Martin EA, Lotspeich-Steininger CA, Koepke JA, tology education series, PN 4206115 A, Hialeah, Fla, 1983,
editors: Clinical hematology: principles, procedures, correlations, Coulter Electronics.
CHAPTER 39 Automated Cell-Counting Instrumentation 623
6. Coulter Electronics: Coulter counter model S-Plus IV with 28. Beckman Coulter: Hematology systems. Available at: http://
three-population differential: product reference manual, PN www.beckman.com. Accessed July 23, 2010.
423560B, Hialeah, Fla, 1983, Coulter Electronics. 29. Sysmex Corporation: Hematology products. Available at:
7. Coulter Electronics: Coulter STKR product reference manual, http://www.sysmex.com Accessed July 23, 2010.
PN 4235547E, Hialeah, Fla, 1988, Coulter Electronics. 30. Beckman Coulter: Gen-S series. Available at: http://
8. TOA Medical Electronics Company: Sysmex model E-5000 www.beckman.com/products/instrument/hematology/
operators manual (CN 461-2104-2), Kobe, Japan, 1985, TOA gens_analyzer.asp. Accessed Sept. 2, 2010.
Medical Electronics Co. 31. Beckman Coulter: Technical Application Information.
9. Coulter Corporation: Coulter STKS operators guide, PN Coulter 3-D VCS technology. Available at: http://www.
423592811, Hialeah, Fla, 1992, Coulter Corp. beckmancoulter.com/products/instrument/hematology/
10. Mohandas N, Clark MR, Kissinger S, et al: Inaccuracies VCS_Technology.pdf Accessed July 23, 2010.
associated with the automated measurement of mean cell 32. Beckman Coulter: LH750. Available at: http://www.
hemoglobin concentration in dehydrated cells. Blood 56: beckmancoulter.com/products/instrument/hematology/
125-128, 1980. lh750_analyzer.asp. Accessed July 22, 2010.
11. Arnfred T, Kristensen SD, Munck V: Coulter counter model 33. Coulter Electronics: Coulter VCS technology casebook, PN
S and model S-plus measurements of mean erythrocyte 4206281-420622A, Hialeah, Fla, 1989, Coulter Electronics.
volume (MCV) are influenced by the mean erythrocyte hae- 34. Beckman Coulter advancements in technology: WBC
moglobin concentration (MCHC). Scand J Clin Lab Invest differential methodology. Available at: http://www.
41:717-721, 1981. beckmancoulter.com/literature/clindiag/DS-12254A%
12. Johnson KL: Basics of flow cytometry. Clin Lab Sci 5:22-24, 20DxH%20WBC.pdf. Accessed September 20, 2009.
1992. 35. Beckman Coulter: Coulter LH 700 series operators guide,
13. Scott R, Sethu P, Harnett CK: Three-dimensional hydrody- Fullerton, Calif, 2003, Beckman Coulter.
namic focusing in a microfluidic Coulter counter. Rev Sci 36. Sysmex Corporation: KX-21N Results you can trust bro-
Instrum 79:046104, 2008. chure. Available at: http://www.sysmex.com/us/515.htm.
14. Rodriquez-Trujillo R, Mills CA, Samitier J, et al: Low cost Accessed September 19, 2009.
micro-Coulter counter with hydrodynamic focusing. Micro- 37. Sysmex Corporation: XE-5000TM Automated Hematology
fluid Nanofluid 3:171-176, 2007. System. Available at: http://www.sysmex.com/us/img/
15. TOA Medical Electronics Company: Sysmex NE-8000 opera- 10_1088_XE5000_01_2008.pdf. Accessed September 18.
tors manual (CN 461-2326-5), Kobe, Japan, 1990, TOA 2009.
Medical Electronics Co. 38. Sysmex Corporation: System XE-2100 operators guide, Kobe,
16. Sysmex Corporation: Sysmex XE-2100 operators manual, Japan, 1999, Sysmex Corp.
Kobe, Japan, 1999, Sysmex Corp. 39. Sysmex Corporation: Identification and monitoring of
17. Sun T: Principles of flow cytometry. In Flow cytometry and functional iron deficiency in ESRD patients using an early
immunohistochemistry for hematologic neoplasms, Philadelphia, erythroid population. Available at: http://www.sysmex.
2008, Wolters Kluwer/Lippincott Williams & Wilkins, com/us/files/10-1098_ret-He_white_paper_2008_web.pdf.
pp 4-14. Accessed August 22, 2009.
18. Jovin TM, Morris SJ, Striker G, et al: Automatic sizing and 40. Abe Y, Wada H, Tomatsu H, et al: A simple technique to
separation of particles by ratios of light scattering intensi- determine thrombopoiesis level using immature platelet
ties. J Histochem Cytochem 24:269-283, 1976. fraction (IPF). Thromb Res 118:463-469, 2006.
19. Ryan DH: Examination of the marrow. In Kaushansky K, 41. Abbott CELL-DYN hematology systems. Available at:
Lichtman MA, Beutler TJ, et al, editors: Williams hematology, http://www.hospitalmanagment.net/contractors/lab/
ed 8, New York, 2010, McGraw-Hill Medical, chap 3. Avail- abbott/. Accessed September 18, 2009.
able at: http://www.accessmedicine.com/content.aspx?aID= 42. Matzdorff AC, Khnel G, Scott S, et al: Comparison of
6106831. Accessed Sept. 2, 2010. flow cytometry and the automated blood analyzer system
20. Ryan DH: Examination of blood cells. In Kaushansky K, CELL-DYN 4000 for platelet analysis. Lab Hematol 4:163-
Lichtman MA, Beutler TJ, et al, editors: Williams hematology, 168, 1998.
ed 8, New York, 2010, McGraw-Hill Medical, chap 2. Avail- 43. Bowden KL, Procopio N, Wystepek E, et al: Platelet clumps,
able at: http://www.accessmedicine.com/content.aspx?aID= nucleated red cells, and leukocyte counts: a comparison
6106433. Accessed Sept. 2, 2010. between the Abbott CELL-DYN 4000 and Coulter STKS. Lab
21. Abbott Laboratories: CELL-DYN 3000 system operators Hematol 4:7-16, 1998.
manual (LN 92420-92401), Abbott Park, Ill, 1993, Abbott 44. Jones RG, Faust A, Glazier J, et al: CELL-DYN 4000: utility
Laboratories. within the core laboratory structure and preliminary com-
22. Abbott Laboratories: CELL-DYN 3500 system operators parison of its expanded differential with the 400-cell manual
manual (LN 92722-92705), Abbott Park, Ill, 1996, Abbott differential count. Lab Hematol 4:34-44, 1998.
Laboratories. 45. Abbott Diagnostics: CELL-DYN rainbow classification program,
23. Siemens Healthcare Diagnostics: ADVIA 120 operators guide, PN 97-9427/R3-20, Abbott Park, Ill, September 1992,
V 3.01.00, Tarrytown, NY, 2009, Siemens Healthcare Abbott Diagnostics.
Diagnostics. 46. Abbott Laboratories: CELL-DYN 3500 system operators
24. Siemens Healthcare Diagnostics: ADVIA 2120/2120i opera- manual: reticulocyte package, (9140293D). Abbott Park, Ill,
tors guide, V4.00.00, Tarrytown, NY, 2008, Siemens Health- 1999, Abbott Laboratories.
care Diagnostics. 47. Abbott Laboratories: CELL-DYN 4000 operators manual,
25. Abbott Diagnostics: Diagnostic products. Available at: Santa Clara, Calif, 1997, Abbott Laboratories.
http://www.abbott.com. Accessed July 23, 2010. 48. Bayer Healthcare: Bayer ADVIA 2120 operators guide,
26. Horiba Medical: ABX hematology: the product line. Avail- Tarrytown, NY, 2005, Bayer Healthcare, 2003.
able at: http://www.horiba.com. Accessed July 23, 2010. 49. Groner W: New developments in flow cytochemistry tech-
27. Siemens Healthcare Diagnostics: Laboratory diagnostics. nology. In Simson E, editor: Proceedings of the technicon H-1
Available at: http://www.medical.siemens.com. Accessed hematology symposium, October 11, 1985, Tarrytown, NY,
July 21, 2010. 1986, Technicon Instruments Corp, pp 1-8.
624 PART VII Cell-Counting Automation

50. Mohandas N, Kim YR, Tycko DH, et al: Accurate and inde- 68. Kessler C, Campbell P, Bolufe V, et al: Immature reticulocyte
pendent measurement of volume and hemoglobin concen- fraction and reticulocyte maturity index. Technical applica-
tration of individual red cells by laser light scattering. Blood tion information. Available at: http://www.beckmancoulter.
68:506-513, 1986. com/literature/ClinDiag/recticliterature.pdf. Accessed Sept.
51. Harris N, Jou JM, Devoto G, et al: Performance evaluation 2, 2010.
of the ADVIA 2120 hematology analyzer: an international 69. Brugnara C, Zurakowski D, DiCanzio J, et al: Reticulocyte
multicenter clinical trial. Lab Hematol 11:62-70, 2005. hemoglobin content to diagnose iron deficiency in children.
52. Koepke J: Current limitations in reticulocyte counting: JAMA 281:2225-2230, 1999.
implications for clinical laboratories. In Porstmann B, 70. Ullrich C, Wu A, Armbsy C, et al: Screening healthy infants
editor: The emerging importance of accurate reticulocyte for iron deficiency using reticulocyte hemoglobin content.
counting, New York, 1993, Caduceus Medical, pp 18-22. JAMA 294:924-930, 2005.
53. Clinical and Laboratory Standards Institute: Methods for 71. Clinical and Laboratory Standards Institute: Performance
reticulocyte counting (flow cytometry and supravital dyes): goals for the internal quality control of multichannel hematology
approved guideline, CLSI document H44-A-2, Wayne, Pa, 2004, analyzers: approved standard, CLSI publication H26-A, Wayne,
Clinical and Laboratory Standards Institute. Pa, 1996, Clinical and Laboratory Standards Institute.
54. College of American Pathologists: CAP surveys: reticulocyte 72. Clinical and Laboratory Standards Institute: Assessment of
(pilot) survey set RT-A, 1993, Northfield, Ill, 1993, College of the clinical accuracy of laboratory tests using receiver operating
American Pathologists. characteristics (ROC) plots: approved guideline, CLSI document
55. College of American Pathologists: CAP surveys: reticulocyte GP1O-A, Wayne, Pa, 1995, Clinical and Laboratory
survey set RT/RT2-A, 2000, Northfield, Ill, 2000, College of Standards Institute.
American Pathologists. 73. Calibration and calibration verification, Clinical Laboratory
56. Coulter Corporation: Introducing a new reticulocyte methodol- Improvement Amendments Brochure No. 3. Available at:
ogy using coulter VCS technology on coulter STKS and MAXM https://www.cms.gov/CLIA/downloads/6065bk.pdf. Accessed
hematology systems, product brochure TC93003201, Miami, Fla, Sept. 2, 2010.
1993, Coulter Corp. 74. Gilmer PR, Williams U, Koepke JA, et al: Calibration
57. TOA Medical Electronics Company: Sysmex R-3000 auto- methods for automated hematology instruments. Am J Clin
mated reticulocyte analyzer: product brochure literature No. Pathol 68:185-190, 1977.
SP-9620, Los Alamitos, Calif, 1991, TOA Medical Electron- 75. Koepke JA: The calibration of automated instruments for
ics Co. accuracy in hemoglobinometry. Am J Clin Pathol 68:180-
58. Coulter Corporation: Coulter STKS with reticulocyte analysis 184, 1977.
operators guide (PN 4237188), Miami, Fla, 1993, Coulter 76. International Committee for Standardization in Haematol-
Corp. ogy: Recommendations for reference method for haemoglo-
59. Beckman Coulter: Technical application information: binometry in human blood (ICSH Standard 1995) and
Coulter VCS reticulocyte method. Available at: http://www. specifications for international haemiglobincyanide stan-
beckmancoulter.com/literature/ClinDiag/ss000126.pdf dard (4th ed). J Clin Pathol 49:271-274, 1996.
Accessed Sept. 2, 2010. 77. Savage RA: Calibration bias and imprecision for automated
60. Ivory K, Sarria B, Fairweather-Tait SJ, et al: Interference with hematology analyzers: an evaluation of the significance of
flow cytometric reticulocyte counts. Int J Lab Hematol short-term bias resulting from calibration of an analyzer
29:352-360, 2007. with S Cal. Am J Clin Pathol 84:186-190, 1985.
61. Bowen KL, Glazier J, Mattson JC: Abbott CELL-DYN 4000 78. College of American Pathologists: Laboratory general: CAP
automated red blood cell analysis compared with routine checklist, 1999 edition, Northfield, Ill, 1998, College of
red blood cell morphology by smear review. Lab Hematol American Pathologists.
4:45-47, 1998. 79. Cornbleet J: Spurious results from automated hematology
62. Colella G: Technical advancements of the H-3 hematology cell counters. Lab Med 14:509-514, 1983.
analyzer. In Baizac F, Chapman S, Verderese C, editors: 80. Tsuda I, Tatsumi N: Evaluation of detection of immature
Conference proceedings: H-3 new perspectives for hematology, WBCs by the new Sysmex SE-9000 automated hematology
New York, 1993, Caduceus Medical, pp 28-29. analyzer. Poster presented at the International Society for
63. Davis BH, Bigelow NC, van Hove L, et al: Evaluation of Laboratory Hematology 7th International Symposium on
automated reticulocyte analysis with immature reticulocyte Technology Innovations in Laboratory Hematology, Lake
fraction as a potential outcomes indicator of anemia in Tahoe, Nev, April 7-10, 1994.
chronic renal failure patients. Lab Hematol 4:169-175, 1998. 81. Bentley SA, Johnson A, Bishop CA: A parallel evaluation of
64. Fourcade C, Jary L, Belaouni H: Reticulocyte analysis pro- four automated hematology analyzers. Am J Clin Pathol
vided by the Coulter GEN-S: significance and interpretation 100:626-632, 1993.
in regenerative and non-regenerative hematologic condi- 82. Miers MK, Exton MG, Hurlbut TA, et al: White blood cell
tions. Lab Hematol 5:153-158, 1999. differentials as performed by the Technicon H-1: evaluation
65. Goldberg M: Recombinant erythropoietin therapy and the and implementation in a tertiary care hospital. Lab Med
detection of iron-deficient erythropoiesis in iron-replete 22:99-106, 1991.
individuals. In Baizac F, Chapman S, Verderese C, editors: 83. Strobel SL, Panke TW, Bills GL: Cold erythrocyte agglutina-
Conference proceedings: H-3 new perspectives for hematology, tion and infectious mononucleosis. Lab Med 24:219-221,
New York, 1993, Caduceus Medical, pp 16-18. 1993.
66. dOnofrio G: Simultaneous H-3 RBC and reticulocyte mea- 84. Duca D, Green R: Red cell distribution width is a poor dis-
surement: is it clinically useful? In Balzac F, Chapman S, criminator for distinguishing iron deficiency anemia from
Verderese C, editors: Conference proceedings: H-3 new perspec- thalassemia minor or the anemia of chronic disease. Poster
tives for hematology, New York, 1993, Caduceus Medical, presented at the 1993 Fall Meeting of the American Society
pp 23-27. of Clinical Pathologists (ASCP) and the College of American
67. Davis BH, Bigelow NC: Automated reticulocyte analysis: Pathologists (CAP), Orlando, Fla, October 16-22, 1993.
clinical practice and associated new parameters. Hematol 85. Green R, King R, Greenbaum A, et al: Early detection of iron
Oncol Clin North Am 8:617-630, 1994. deficiency by direct measurement of red cell hemoglobin
CHAPTER 39 Automated Cell-Counting Instrumentation 625
concentration: sequential studies in phlebotomized normal fluorescence flow cytometry, the Abbott CELL-DYN 4000
volunteers. Blood 76(suppl):122, 1990. hematology analyzer, and microscopy. Lab Hematol 4:64-
86. Mohandas N, Greenbaum A, Green R: Variability in cell 70, 1998.
hemoglobin content of individual red cells can be used to 107. Clinical Laboratory and Standards Institute: Reference
discriminate iron deficiency from thalassemia minor. Blood leukocyte differential count (proportional) and evaluation of
76:155, 1990. instrumental methods: approved standard, CLSI document H20-
87. Brugnara C, Chambers LA, Malynn E, et al: Red blood cell A, Wayne, Pa, 1992, Clinical Laboratory and Standards
regeneration induced by subcutaneous recombinant eryth- Institute.
ropoietin: iron-deficient erythropoiesis in iron-replete sub- 108. Warner BA, Reardon DM, Marshall DP: Automated haema-
jects. Blood 81:956-964, 1993. tology analysers: a four-way comparison. Med Lab Sci
88. Coulter Electronics: Proceedings of the coulter automated dif- 47:285-296, 1990.
ferential international symposium, September 26, 1986, Hialeah, 109. Swaim WR: Laboratory and clinical evaluation of white
Fla, 1987, Coulter Electronics. blood cell differential counts: comparison of the Coulter
89. Reardon DM, Hutchinson D, Preston FE, et al: The routine VCS, Technicon H-1, and 800-cell manual method. Am J
measurement of platelet volume: a comparison of aperture- Clin Pathol 95:381-388, 1991.
impedance and flow cytometric systems. Clin Lab Haematol 110. Buttarello M, Gadotti M, Lorenz C, et al: Evaluation of four
7:251-257, 1985. automated hematology analyzers: a comparative study of
90. Allen JK, Batjer ID: Evaluation of an automated method for differential counts (imprecision and inaccuracy). Am J Clin
leukocyte differential counts based on electronic volume Pathol 97:345-352, 1992.
analysis. Arch Pathol Lab Med 109:534-539, 1985. 111. Goossens W, Hove LV, Verwilghen RL: Monocyte counting:
91. Pierre RV, Payne BA, Lee WK, et al: Comparison of four discrepancies in results obtained with different automated
leukocyte differential methods with the National Commit- instruments. J Clin Pathol 44:224-227, 1991.
tee for Clinical Laboratory Standards (NCCLS) reference 112. Seaberg R, Cuomo J: Assessment of monocyte counts
method. Am J Clin Pathol 87:201-209, 1987. derived from automated instrumentation. Lab Med 24:222-
92. Payne BA, Pierre RV, Lee WK: Evaluation of the TOA E-5000 224, 1993.
automated hematology analyzer. Am J Clin Pathol 88:51-57, 113. Watson JS, Ross DW: Characterization of myelodysplastic
1987. syndromes by flow cytochemistry with the Technicon H-1.
93. Ross DW, Watson JS, Davis PH, et al: Evaluation of the J Med Tech 4:18-20, 1987.
Coulter three-part differential screen. Am J Clin Pathol 114. Krause JR, Costello RT, Krause J, et al: Use of the Technicon
84:481-484, 1985. H-1 in the characterization of leukemias. Arch Pathol Lab
94. Cornbleet J, Kessinger S: Evaluation of Coulter S-Plus three- Med 112:889-894, 1988.
part differential in a population with a high prevalence of 115. Penchansky L, Krause JR: Flow cytochemical study of acute
abnormalities. Am J Clin Pathol 84:620-626, 1985. leukemia of childhood with the Technicon H-1. Lab Med
95. Miers MK, Fogo AB, Federspiel CF, et al: Evaluation of the 22:184-189, 1991.
Coulter S-Plus IV differential as a screening tool in a tertiary 116. Bessman D, Williams U, Gilmer PR: Mean platelet volume:
care hospital. Am J Clin Pathol 87:745-751, 1987. the inverse relation of platelet size and count in normal
96. Ross DW, Bentley SA: Evaluation of an automated hematol- subjects and an artifact of other particles. Am J Clin Pathol
ogy system (Technicon H-1). Arch Pathol Lab Med 110:803- 76:289-293, 1981.
808, 1986. 117. Walters JG, Garrity PF: Case studies in the new morphology,
97. Bollinger PB, Drewinko B, Brailas CD, et al: The Technicon McGraw Park, Ill, 1987, American Scientific Products.
H-1: an automated hematology analyzer for today and 118. Simson E, Ross DW, Kocher WD: Atlas of automated
tomorrow. Am J Clin Pathol 87:71-78, 1987. cytochemical hematology, Tarrytown, NY, 1988, Technicon
98. Watson JS, Davis RA: Evaluation of the Technicon H-1 Instruments Corp.
hematology system. Lab Med 18:316-322, 1987. 119. Hinchliffe RF, Helliwell MM: Red cells in children: an H-1
99. Warner BA, Reardon DM: A field evaluation of the Coulter atlas, Tarrytown, NY, 1993, Bayer Diagnostics.
STKS. Am J Clin Pathol 95:207-217, 1991. 120. Beckman Coulter: Coulter Gen-S system: clinical case studies,
100. Hallawell R, OMalley C, Hussein S, et al: An evaluation of PN 9914978, Brea, Calif, 1996, Beckman Coulter.
the Sysmex NE-8000 hematology analyzer. Am J Clin Pathol 121. Sysmex Corporation: Products/Hematology available at
96:594-601, 1991. http://www.sysmex.com/us/515.htm. Accessed Sept. 2, 2010.
101. Cornbleet PJ, Myrick D, Judkins S, et al: Evaluation of the 122. Harris N, Kunicka J, Kratz A: The ADVIA 2120 hematology
CELL-DYN 3000 differential. Am J Clin Pathol 98:603-614, system: flow cytometry-based analysis of blood and body
1992. fluids in the routine hematology laboratory. Lab Hematol
102. Cornbleet PJ, Myrick D, Levy R: Evaluation of the Coulter 11:47-61, 2005.
STKS five-part differential. Am J Clin Pathol 99:72-81, 1993. 123. Beckman Coulter: Body fluid analysis made simpleand
103. Brigden ML, Page NE, Graydon C: Evaluation of the Sysmex automated. Available at: http://www.beckmancoulter.com/
NE-8000 automated hematology analyzer in a high-volume products/instrument/hematology/body-fluid-application.
outpatient laboratory. Am J Clin Pathol 100:618-625, 1993. asp. Accessed Sept. 2, 2010.
104. Rumke CL: The statistically expected variability in differen- 124. Camden TL: How to select the ideal hematology analyzer.
tial leukocyte counting. In Koepke JA, editor: Differential MLO Med Lab Obs 25:29-33, 1993.
leukocyte counting, Skokie, Ill, 1978, College of American 125. van Hove L: Guest editorial: which hematology analyzer do
Pathologists. you need? Lab Hematol 4:32-33, 1998.
105. Koepke JA, Dotson MA, Shifman MA: A critical evaluation 126. Albitar M, Dong Q, Saunder D, et al: Evaluation of auto-
of the manual/visual differential leukocyte counting mated leukocyte differential counts in a cancer center. Lab
method. Blood Cells 11:173-181, 1985. Hematol 5:10-14, 1999.
106. Paterakis G, Kossivas L, Kendall R, et al: Comparative evalu- 127. Culp NB, Fritsma G: New approaches to nucleated RBC
ation of the erythroblast count generated by three-color correction of WBC counts. Clin Lab Sci 3(4):239, 1990.
PART VIII Hemostasis and Thrombosis

40 Normal Hemostasis and


Coagulation
Margaret G. Fritsma and George A. Fritsma

OUTLINE OBJECTIVES
Overview of Hemostasis After completion of this chapter, the reader will be able to:
Primary Hemostasis
Secondary Hemostasis 1. List the systems that interact to provide hemostasis. 7. Diagram fibrinogen structure, fibrin formation, fibrin
Fibrinolysis 2. Describe the properties of the vascular intima in polymerization, and fibrin cross-linking.
Vascular Intima in the initiation and regulation of hemostasis and 8. Distinguish between serine proteases and cofactors
Hemostasis fibrinolysis. in the coagulation pathway.
Anticoagulant Properties 3. List the hemostatic functions of tissue factorbearing 9. Describe the tissue factor pathway, amplification
of Intact Vascular Intima cells and blood cells, especially platelets, in pathway, and contact activation in coagulation.
Procoagulant Properties hemostasis. 10. List the factors in order of reaction in the extrinsic,
of Damaged Vascular 4. Describe the relationships among platelet function, intrinsic, and common pathways.
Intima von Willebrand factor, and fibrinogen and their 11. Describe the in vivo coagulation process and the role
Fibrinolytic Properties of
impact on hemostasis. of tissue factorbearing cells and platelets.
Vascular Intima
5. Describe the nature, origin, and function of each of 12. Show how tissue factor pathway inhibitor, the protein
Platelets
Tissue FactorBearing the tissue and plasma factors necessary for normal C pathway, and the serine protease inhibitors
Cells, Erythrocytes, coagulation. function to regulate coagulation and prevent
Monocytes, and 6. Explain the role of vitamin K in the production and thrombosis.
Lymphocytes function of plasma clotting factors in the prothrombin 13. Describe the fibrinolytic pathway, its regulators, and
Coagulation System group. its products.
Nomenclature of
Procoagulants
Classification of
Procoagulants
Coagulation Pathways
Cell-Based (In Vivo) CASE STUDY
Coagulation After studying the material in this chapter, the reader should be able to respond to the following
Coagulation Regulatory case study:
Mechanisms
Tissue Factor Pathway A pregnant woman developed a blood clot in her left leg (deep vein thrombosis, or DVT). Her
Inhibitor mother reportedly had a history of thrombophlebitis. She had a brother who was diagnosed with
Protein C Regulatory a DVT following a flight from Los Angeles to Sydney, Australia.
System
1. Is this hemostatic disorder typical of an acquired or an inherited condition?
Serine Protease Inhibitors
2. Are these symptoms most likely caused by a deficiency of a procoagulant or an inhibitor?
(Serpins)
Fibrinolysis
Plasminogen and Plasmin
Plasminogen Activation
and Control
Fibrin Degradation
Products and D-Dimer

626
CHAPTER 40 Normal Hemostasis and Coagulation 627

P
hysiologic hemostasis is an amazing and complex plasma transports at least 16 glycoproteins, mostly trypsinlike
process that keeps blood fluid in the circulation and enzymes called serine proteases. These proteins circulate as inac-
then, when an injury occurs, produces a clot to stop tive zymogens that become activated during the process of
the bleeding, keeps the clot confined to the site of injury, coagulation and, in turn, form complexes that activate other
and, finally, dissolves the clot as the wound heals. When hemo- zymogens to ultimately generate thrombin, an enzyme that
stasis systems are out of balance, thrombosis (clotting) or converts fibrinogen to a localized fibrin clot. Some coagulation
hemorrhage (bleeding) can be life-threatening. Medical labora- proteins are cofactors that join in forming a complex and func-
tory scientists and hematologists investigate all of the major tion to stabilize and enhance enzymatic action. Still others are
hemostasis systemsthe blood vessels, platelets, and plasma control proteins that regulate each step of the process and prevent
proteinsto prevent, predict, diagnose, and manage hemo- excessive thrombin generation. Figure 40-1 shows a simplified
static disease. sequence of activation of the coagulation proteins to form
fibrin. Secondary hemostasis is triggered by tissue damage that
exposes TF, a protein present in the membrane of subendothe-
OVERVIEW OF HEMOSTASIS
lial cellsfibroblasts and smooth muscle cellsbut not on
The cellular elements of hemostasis are primarily the vascular endothelial cells. Exposed TF forms a complex with activated
intima, extravascular tissue factor (TF)bearing cells, and plate- factor VII, which activates factors IX and X. Factor IX combines
lets. The plasma components are the coagulation and fibrino- with its cofactor, factor VIII, also to activate factor X. The complex
lytic proteins and their inhibitors. formed by factor X and its cofactor, factor V, converts prothrom-
bin to thrombin. Thrombin is the key enzyme in coagulation.
Primary Hemostasis It splits peptides from fibrinogen to produce fibrin, activates
Hemostasis involves the interaction of vasoconstriction, plate- factor XIII to cross-link and stabilize the clot, activates platelets,
let aggregation, and coagulation enzymes to stop bleeding and feeds back to activate factors V, VIII, and XI. Fibrin forma-
(Table 40-1). Primary hemostasis refers to the role of blood tion in secondary hemostasis is necessary to control bleeding,
vessels and platelets in the initial formation of a platelet plug especially from large wounds incurred through trauma, surgery,
in response to a vascular injury. Primary hemostasis mecha- or dental procedures. The coagulation system, similar to other
nisms are activated either by injuries to blood vessels or by the humoral amplification mechanisms, is complex because it
commonplace desquamation of dying or damaged endothelial translates a diminutive physical or chemical stimulus into a
cells. In primary hemostasis, the blood vessel contracts to seal profound lifesaving event.1 The absence of a single plasma pro-
the wound or reduce the blood flow (vasoconstriction). Pro- coagulant may doom the individual to lifelong anatomic hemor-
coagulant substances are exposed or released by the damaged rhage, chronic inflammation, and transfusion dependence.
endothelium. Platelets are activated, adhere to exposed colla-
gen, secrete the contents of their granules, and aggregate with
Intrinsic
other platelets to form a platelet plug. Vasoconstriction and
platelet plug formation are the initial, rapid, short-lived Vlla Xla Xlla
Extrinsic
response to vessel damage, but by themselves they are inade- Pre-K HMWK
TF
quate to control major bleeding in the long term. Eventually
the plug must be reinforced by fibrin. Defects in the primary IXa Vllla
hemostasis system such as thrombocytopenia, platelet disor-
ders, or von Willebrand disease can cause debilitating, some-
times fatal, chronic hemorrhage. Common Xa Va

Secondary Hemostasis
Secondary hemostasis is the activation of a series of plasma pro-
Thr Xllla
teins in the coagulation system to form a fibrin clot. Blood

Cross-linked
TABLE 40-1 Primary and Secondary Hemostasis Fibrinogen Fibrin polymer
fibrin

Primary Hemostasis Secondary Hemostasis Figure 40-1 Simplified representation of the coagulation pathway.
Exposed tissue factor (TF) activates factor VII, which activates factors IX and
Activated by desquamation and Activated by large injuries to blood X. Factor IX:VIII complex also activates X, and the factor Xa:Va complex
small injuries to blood vessels vessels and surrounding tissues activates prothrombin (factor II). The resulting thrombin (factor IIa) cleaves
Involves vascular intima and Involves platelets and coagulation fibrinogen to form fibrin polymer and cross-linked fibrin. In vitro exposure to
platelets system negatively charged surfaces activates the contact factors XII, pre-K and high-
Rapid, short-lived response Delayed, long-term response molecular-weight kininogen (HMWK), which proceed to activate factors XI,
Procoagulant substances exposed Tissue factor exposed on cell IX, VIII, X, V, II, and I, the intrinsic pathway. The extrinsic pathway factors are
or released by damaged or membranes VII, X, V, II, and I. Thrombin also activates factors V, VIII, XI, and XIII. Coagula-
activated endothelial cells tion complexes form on TF-bearing cells and on platelet phospholipid mem-
branes in the presence of Ca2+.
628 PART VIII Hemostasis and Thrombosis

Fibrinolysis inner surface that evens the blood flow and prevents harmful
The final event of hemostasis is fibrinolysis, the slow digestion turbulence. Endothelial cells form a physical barrier between
and removal of the fibrin clot as healing occurs.2 Plasminogen blood and the substances in the basement membrane that
is bound to fibrin during coagulation and is activated by tissue activate clotting, such as collagen, which promotes platelet
plasminogen activator (TPA) into the serine protease, plasmin. activation and adhesion, and TF, which activates coagulation
Plasmin slowly and systematically degrades the fibrin clot into and fibrin formation. Endothelial cells synthesize and secrete
fragments called fibrin degradation products, designated X, Y, D, a variety of substances that maintain normal blood flow.
E, and D-dimer. Prostacyclin, a platelet inhibitor, is synthesized through the
Although the vascular intima and platelets usually are asso- eicosanoid pathway (see Chapter 13) and prevents unnecessary
ciated with primary hemostasis, and coagulation and fibrino- or undesirable platelet activation in intact vessels.5 Nitric oxide
lysis are associated with secondary hemostasis, all systems is synthesized in endothelial cells, vascular smooth muscle
interact in early and late hemostatic events. For example, plate- cells, neutrophils, and macrophages. Nitric oxide counteracts
lets, although a key component of primary hemostasis, also vasoconstriction and maintains healthy arterioles.6 Another
secrete some coagulation factors stored in their granules (V, important endothelial cell anticoagulant is tissue factor pathway
VIII, fibrinogen, von Willebrand factor [VWF]) and, very inhibitor (TFPI), which controls the TF or extrinsic coagulation
importantly, provide a phospholipid cell surface on which pathway. Finally, endothelial cells synthesize and secrete onto
coagulation complex formation can take place. The remainder their cell surfaces two inhibitors of thrombin formation, throm-
of this chapter examines vascular intima, platelets, normal bomodulin and heparan sulfate. Thrombomodulin is a mem-
coagulation, coagulation control, and fibrinolysis in detail. brane protein that activates the protein C pathway. The protein
C pathway downregulates coagulation by digesting activated
factors V and VIII, thereby inhibiting fibrin formation. Heparan
VASCULAR INTIMA IN HEMOSTASIS
sulfate is a glycosaminoglycan that retards coagulation by acti-
Intimal cells and their environment play a premier role in vating antithrombin, a coagulation regulatory protein.7 The
hemostasis.3 The innermost lining of blood vessels is a mono- pharmaceutical heparin, manufactured from porcine gut tissues,
layer of metabolically active endothelial cells (Box 40-1, Figure
40-2).4 These form a smooth, unbroken surface that promotes
the fluid passage of blood and prevents turbulence that other-
wise may activate platelets and coagulation enzymes. An
SMC FB SMC FB SMC
elastin-rich internal elastic lamina and its surrounding layer of
connective tissues support the endothelial cells. In all blood IEL
vessels, fibroblasts occupy the connective tissue layer and EC EC EC EC EC EC EC EC
produce collagen. Smooth muscle cells in arteries and arteri-
RBC
oles, but not in the walls of veins, venules, or capillaries, con- RBC PLT RBC
PLT
PLT
tract during primary hemostasis.
EC EC EC EC EC EC EC EC
Anticoagulant Properties of IEL
Intact Vascular Intima
Normally, the intact vascular endothelium prevents bleeding SMC FB SMC FB SMC
and thrombosis by inhibiting platelet and coagulation activa-
tion and by promoting fibrinolysis. Intravascular thrombosis
Figure 40-2 Normal blood flow in intact vessels. Smooth, rhomboid
is prevented by several mechanisms (Box 40-2). First, endothe-
endothelial surfaces promote even flow. Larger cells are focused toward the
lial cells are rhomboid and contiguous, providing a smooth center. The endothelium provides several hemostasis-suppressing materials.
EC, Endothelial cell; FB, fibroblast; IEL, internal elastic lamina; PLT, platelet;
RBC, red blood cell; SMC, smooth muscle cell; lines indicate collagen.
BOX 40-1 Vascular Intima

Innermost Vascular Lining BOX 40-2 Anticoagulant Properties of Intact


Endothelial cells (endothelium) Endothelium
Composed of rhomboid cells presenting a smooth, contiguous surface
Supporting the Endothelial Cells Secretes the eicosanoid platelet inhibitor prostacyclin
Internal elastic lamina, composed of elastin and collagen, surrounds Secretes vascular relaxing factor nitric oxide
endothelium Secretes the anticoagulant glycosaminoglycan heparan sulfate
Secretes coagulation extrinsic pathway regulator tissue factor pathway
Subendothelial Connective Tissue inhibitor
Collagen and fibroblasts in veins Maintains cell membrane thrombomodulin, a protein C coagulation
Collagen, fibroblasts, and smooth muscle cells in arteries control system activator
CHAPTER 40 Normal Hemostasis and Coagulation 629

resembles heparan sulfate in its antithrombin activity. Heparin plasminogen to plasmin, which slowly digests fibrin and
is used extensively as a therapeutic agent to prevent propaga- restores blood flow. Endothelial cells, as well as other cells, may
tion of the thrombi that cause coronary thrombosis, strokes, secrete plasminogen activator inhibitor 1 (PAI-1), a TPA control
deep vein thromboses, and pulmonary emboli.8 protein that inhibits fibrinolysis.14 Thrombin bound to throm-
bomodulin activates thrombin-activatable fibrinolysis inhibitor
Procoagulant Properties of Damaged (TAFI), which increases the tendency for thrombus formation.
Vascular Intima Although the significance of the vascular intima in hemo-
Although the intact endothelium has anticoagulant properties, stasis is well recognized, there are few valid laboratory methods
when damaged, the vascular intima promotes coagulation. to assess the integrity of endothelial cells, smooth muscle cells,
First, any harmful local stimulus, be it mechanical or chemical, fibroblasts, and their collagen matrix.15 The diagnosis of blood
induces vasoconstriction in arteries and arterioles (Table 40-2). vessel disorders is often based on clinical symptoms, family
Smooth muscle cells contract, the vascular lumen narrows or history, and laboratory tests that rule out platelet or coagula-
closes, and blood flow to the injured site is minimized. tion disorders.
Although veins and capillaries do not have smooth muscle
cells, bleeding into tissues creates some extravascular pressure
PLATELETS
on the blood vessel as well. Second, the subendothelial con-
nective tissues of arteries and veins are rich in collagen, a flex- Platelets are produced from the cytoplasm of bone marrow
ible, elastic structural protein that binds and activates platelets. megakaryocytes (see Chapter 13).16 Although platelets are only
Some connective tissue degeneration occurs naturally in aging, 2 to 3m in diameter on a fixed, stained peripheral blood film,
which leads to an increased tendency toward bruising. Third, they are complex, metabolically active cells that interact with
endothelial cells secrete VWF, a 600,000- to 20,000,000-D gly- their environment and initiate and control hemostasis.17
coprotein that is necessary for platelets to adhere to exposed In an injury, platelets adhere, aggregate, and secrete the
subendothelial collagen in arterioles.9 Fourth, on activation, contents of their granules (Table 40-3).18,19 Adhesion is the
endothelial cells secrete and coat themselves with P-selectin, property of binding to nonplatelet surfaces such as subendo-
an adhesion molecule that promotes platelet and leukocyte thelial collagen (Figures 40-3 and 40-4). VWF links platelets to
binding.10 Endothelial cells also secrete immunoglobulin-like collagen in areas of high shear stress such as arteries and arte-
adhesion molecules called intercellular adhesion molecules rioles, whereas platelets may bind directly to collagen in
(ICAMs) and platelet endothelial cell adhesion molecules (PECAMs) damaged veins and capillaries. VWF binds platelets through the
that promote leukocyte binding.11 Finally, subendothelial cells glycoprotein (GP) Ib/IX/V receptor. The importance of platelet
(i.e., smooth muscle cells and fibroblasts) support the constitu- adhesion is underscored by bleeding disorders such as Bernard-
tive surface protein TF.12 Exposed TF activates the coagulation Soulier syndrome in which the GP Ib/IX/V receptor is absent,
system through factor VII. TF also appears on the surface and von Willebrand disease, in which VWF is missing or defec-
of endothelial cells and on bloodborne monocytes during tive. Aggregation is the attachment of platelets to each other
inflammation.13 (Figure 40-5).20 When platelets are activated, a change in the
GP IIb/IIIa receptor allows binding of fibrinogen, as well as
Fibrinolytic Properties of Vascular Intima VWF and fibronectin. Fibrinogen binds to GP IIb/IIIa receptors
Endothelial cells support fibrinolysis with the secretion of TPA. on adjacent platelets and cross-links them in the presence of
During thrombus formation, both TPA and plasminogen bind Ca2+. Fibrinogen binding is essential for platelet aggregation,
to polymerized fibrin. TPA activates fibrinolysis by converting as evidenced by bleeding and compromised aggregation in
patients with afibrinogenemia or in patients who lack GP
IIb/IIIa (Glanzmann thrombasthenia). In in vitro platelet
TABLE 40-2 Procoagulant Properties of the Damaged
Vascular Intima
Structure Procoagulant Property TABLE 40-3 Platelet Function
Smooth muscle cells in arterioles and Induce vasoconstriction Function Characteristics
arteries
Exposed subendothelial collagen Binds VWF and platelets Adhesion: platelets roll and Reversible; seals endothelial gaps,
Damaged or activated endothelial cells Secrete VWF cling to nonplatelet surfaces some secretion of growth factors,
Secrete adhesion molecules: in arterioles von Willebrand factor
P-selectin, ICAMs, PECAMs is necessary for adhesion
Exposed smooth muscle cells and Tissue factor exposed on cell Aggregation: platelets adhere to Irreversible; platelet plugs form,
fibroblasts membranes each other platelet contents are secreted,
Endothelial cells in inflammation Tissue factor is induced by requires fibrinogen
inflammation Secretion: platelets discharge Irreversible; occurs during
the contents of their granules aggregation, platelet contents are
ICAMs, Intercellular adhesion molecules; PECAMs, platelet endothelial cell adhesion secreted, essential to coagulation
molecules; VWF, von Willebrand factor.
630 PART VIII Hemostasis and Thrombosis

TABLE 40-4 Platelet Granule Contents


SMC FB SMC FB SMC Platelet -Granules
Platelet -Granules (Dense Bodies)
IEL
PLT PLT PLT
EC EC EC EC EC EC Large molecules Small molecules
-Thromboglobulin Adenosine diphosphate (activates
RBC
RBC PLT RBC Factor V neighboring platelets)
PLT
PLT Factor XI Adenosine triphosphate
Protein S Calcium
EC EC EC EC EC EC EC EC
Fibrinogen Serotonin (vasoconstrictor)
IEL
Von Willebrand factor
SMC FB SMC FB SMC Platelet factor 4 (heparin inhibitor)
Platelet-derived growth factor

Figure 40-3 Platelet adhesion. On desquamation of endothelial cells


(ECs), platelets (PLTs) adhere to the subendothelium and fill in until new
endothelial cells grow. FB, Fibroblast; IEL, internal elastic lamina; RBC, red
blood cell; SMC, smooth muscle cell; lines indicate collagen. aggregation studies, the most commonly used agonists to
induce aggregation are thrombin, arachidonic acid, adenosine
diphosphate (ADP), collagen, and epinephrine, which bind to
receptors on the platelet membrane. Platelets secrete the con-
tents of their granules during adhesion and aggregation,
PLT although most secretion occurs late in the platelet activation
Platelet membrane process. Platelets secrete procoagulants, such as VWF, factor V,
integrin receptor site Platelet binding ligand and fibrinogen, as well as control proteins, Ca2+, ADP, and
Collagen binding VWF other hemostatic molecules. See Table 40-4 for a summary of
site the contents of platelet -granules and dense bodies.
EC EC During activation, ADP and Ca2+ activate phospholipase A2,
Basement which converts membrane phospholipid to arachidonic acid.
membrane Cyclooxygenase converts arachidonic acid into prostaglandin
FB SMC endoperoxides. In the platelet, thromboxane synthetase con-
verts prostaglandins into thromboxane A2, which causes Ca2+
to be released and promotes platelet aggregation and vasocon-
Figure 40-4 Interaction of platelets (PLTs), von Willebrand factor (VWF), striction (Figure 40-6). Aspirin permanently inactivates cyclo-
and collagen. In arterioles and arteries, where blood moves rapidly, platelets oxygenase, blocking thromboxane A2 production and causing
adhere by binding VWF. The larger VWF multimers form a fibrillar carpet on impairment of platelet function (aspirin effect).
which the platelets assemble. EC, Endothelial cell; FB, fibroblast; SMC, See Chapter 13 for an in-depth description of platelet struc-
smooth muscle cell; lines indicate collagen. ture and function. Platelet disorders are considered in detail in
Chapters 43 and 44.
The platelet membrane is the key surface for coagulation
enzyme-cofactor-substrate complex formation. Platelets supply
both phospholipid (phosphatidylserine) and Ca2+, as well as
SMC FB SMC FB SMC procoagulant factors and receptors. Coagulation is initiated on
TF-bearing cells with the formation of the complex TF:VIIa,
IEL which activates IX and X and produces enough thrombin to
EC PLT PLT PLT PLT
EC EC EC EC
PLT PLT
EC activate platelets and factors V, VIII, and XI. Once platelets are
PLT PLT PLT
activated, coagulation proceeds with the formation of tenase
RBC PLT RBC (IXa:VIIIa) and prothrombinase (Xa:Va) on the platelet
RBC PLT
PLT surface, ultimately generating a burst of thrombin at the site of
EC EC EC EC EC EC EC EC injury. See subsequent text for more detail.
IEL

SMC FB SMC FB SMC TISSUE FACTORBEARING CELLS,


ERYTHROCYTES, MONOCYTES,
AND LYMPHOCYTES
Figure 40-5 Platelet aggregation. In severe vascular damage, platelets
(PLTs) aggregate to form a platelet plug. EC, Endothelial cell; FB, fibroblast; TF is an intrinsic transmembrane receptor found on extravas-
IEL, internal elastic lamina; RBC, red blood cell; SMC, smooth muscle cell; cular cells such as fibroblasts, smooth muscle cells, and peri-
lines indicate collagen. cytes but, under normal conditions, not on vessel endothelial
CHAPTER 40 Normal Hemostasis and Coagulation 631

Agonist binds
membrane receptor
Ph
osp
ho
lipa
se
A
Aspirin acetylates 2
COX 1 and reduces
TXA2 production

Arachidonic acid
COOH
Cyclooxygenase-1

PGG2/PGH2

TXA Synthase

Thromboxane A2
Ca2 release
Platelet aggregation
Vasoconstriction

Thromboxane B2 is a stable,
measurable plasma product that
is reduced in aspirin therapy

Figure 40-6 Arachidonic acid and aspirin effect. Phospholipase A2 converts membrane phospholipids into arachidonic acid during platelet activation.
Arachidonic acid is converted to prostaglandin endoperoxides (PGG2 /PGH2) by cyclooxygenase, then to thromboxane A2 (TxA2). TxA2 causes release of Ca2+,
which promotes platelet aggregation and vasoconstriction. Aspirin permanently blocks the action of cyclooxygenase-1 and TxA2 synthesis, impairing platelet
function.

cells.21 Vessel injury exposes blood to the subendothelial procoagulant becomes activated, a lower-case a appears behind
TF-bearing cells and leads to activation of coagulation through the numeral; for instance, activated factor VII is VIIa. Both
factor VIIa. TF is also expressed in high levels in the brain, lung, zymogens and cofactors become activated in the coagulation
placenta, heart, kidney, and testis. process.
Erythrocytes, monocytes, and lymphocytes also participate We customarily call factor I fibrinogen and factor II prothrom-
in hemostasis. Erythrocytes add bulk and structural integrity to bin, although occasionally they are identified by their numer-
the fibrin clot; there is a tendency to bleed in anemia. In als. The numeral III was given to tissue thromboplastin, a crude
inflammatory conditions, monocytes and lymphocytes, as well mixture of TF and phospholipid. Now that the precise structure
as endothelial cells, provide surface-borne TF that triggers coag- of TF has been described, the numeral designation is seldom
ulation. Leukocytes also have a series of membrane integrins used. The numeral IV identified the plasma cation calcium
and selectins that bind adhesion molecules and help stimulate (Ca2+); however, calcium is referred to by its name or chemical
the production of inflammatory materials that promote the symbol, not by its numeral. The numeral VI was assigned to a
wound healing process.22 procoagulant that later was determined to be activated factor
V; VI was withdrawn from the naming system and never reas-
signed. Factor VIII, antihemophilic factor, is a cofactor that
COAGULATION SYSTEM
circulates linked to a large carrier protein, VWF. Prekallikrein,
Nomenclature of Procoagulants also called Fletcher factor, and high-molecular-weight kinino-
Plasma transports at least 16 procoagulants, also called coagula- gen (HMWK), also called Fitzgerald factor, have never received
tion factors or clotting factors. Nearly all are glycoproteins Roman numerals because they belong to the kallikrein and
synthesized in the liver, although a few are made by mono- kinin systems, and their primary functions lie within these
cytes, endothelial cells, and megakaryocytes (Table 40-5, Figure systems. Platelet phospholipids, particularly phosphatidylser-
40-7). Eight are enzymes that circulate in an inactive form ine, are required for the coagulation process but were given no
called zymogens. Others are cofactors that bind and stabilize Roman numeral; instead they were called collectively platelet
their respective enzymes. During clotting, the procoagulants factor 3. The molecular weights, plasma concentrations, and
become activated and produce a localized thrombus. Addi- plasma half-lives of the procoagulants are given in Table 40-5.
tional plasma glycoproteins are controls that regulate the coag- These essential pieces of clinical information assist in the
ulation process. interpretation of laboratory tests, monitoring of anticoagulant
In 1958 the International Committee for the Standardiza- therapy, and design of effective replacement therapies in
tion of the Nomenclature of the Blood Clotting Factors offi- deficiency-related hemorrhagic diseases. For example, factor
cially named the plasma procoagulants using Roman numerals VIII has a short half-life of 12 hours, so replacement therapy
in the order of their initial description or discovery.23 When a for hemophilic individuals who are deficient in factor VIII is
632 PART VIII Hemostasis and Thrombosis

TABLE 40-5 Plasma Procoagulants: Function, Molecular Weight, Plasma Half-Life, and Plasma Concentration
Molecular Weight Half-Life Mean Plasma
Factor Customary Name Function (daltons) (hours) Concentration
I* Fibrinogen Thrombin substrate, polymerizes to 340,000 100-150 200-400mg/dL
form fibrin
II* Prothrombin Serine protease 71,600 60 10mg/dL
III* Tissue factor Cofactor 44,000 Insoluble None
IV* Ionic calcium Mineral 40 NA 8-10mg/dL
V Labile factor Cofactor 330,000 24 1mg/dL
VII Stable factor Serine protease 50,000 6 0.05mg/dL
VIII Antihemophilic factor Cofactor 260,000 12 0.01mg/dL
VWF von Willebrand factor Factor VIII carrier and platelet adhesion 600,000-20,000,000 24 1mg/dL
IX Christmas factor Serine protease 57,000 24 0.3mg/dL
X Stuart-Prower factor Serine protease 58,800 48-52 1mg/dL
XI Plasma thromboplastin Serine protease 143,000 48-84 0.5mg/dL
antecedent (PTA)
XII Hageman factor Serine protease 84,000 48-70 3mg/dL
Prekallikrein Fletcher factor, pre-K Serine protease 85,000 35 35-50mcg/mL
High-molecular-weight Fitzgerald factor, HMWK Cofactor 120,000 156 5mg/dL
kininogen
XIII Fibrin-stabilizing factor (FSF) Transglutaminase, transamidase 320,000 150 2mg/dL
Platelet factor 3 Phospholipids, Assembly molecule Released by
phosphatidylserine, PF3 platelets

From Greenberg DL, Davie EW: The blood coagulation factors: their complementary DNAs, genes, and expression. In Colman RW, Marder VJ, Clowes, AM, et al, editors: Hemo-
stasis and thrombosis: basic principles and clinical practice, ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins, pp 21-58.
*These factors are customarily identified by name rather than Roman numeral.

Protein Z
VIII

XII HMWK V
Prekallikrein X

VII Cofactors
XI Zymogens

IX Tissue factor Protein S


Prothrombin
XIII

Thrombomodulin

Heparin Protein C
cofactor II Antithrombin

Control proteins Fibrinogen substrate


TFPI

a2-macroglobulin
a1-antitrypsin

ZPI

Figure 40-7 Plasma procoagulants, cofactors, and anticoagulants. HMWK, High-molecular-weight kininogen; TFPI, tissue factor pathway inhibitor; ZPI,
protein Zdependent protease inhibitor.
CHAPTER 40 Normal Hemostasis and Coagulation 633

TABLE 40-6 Plasma Procoagulant Serine Proteases TABLE 40-7 Plasma Procoagulant Cofactors
Inactive Active Inactive Form Active Form Binds
Zymogen Protease Cofactor Substrate
Tissue factor Exposed tissue factor VIIa
Prothrombin Thrombin Fibrinogen, V, V Va Xa
(II) (IIa) VIII, XI, XIII VIII VIIIa IXa
VII VIIa Tissue factor IX, X High-molecular-weight Kinin XIIa, Prekallikrein
IX IXa VIIIa X kininogen
X Xa Va Prothrombin
XI XIa IX
XII XIIa High-molecular-weight XI
TABLE 40-8 Vitamin KDependent Coagulation Factors
kininogen
Prekallikrein Kallikrein High-molecular-weight XI Procoagulants Regulatory Proteins
kininogen Prothrombin (II) Protein C
VII Protein S
IX Protein Z
administered every 12 hours. For most factors, the level of X
hemostatic effectiveness is 25% to 30%. This is the minimum
level that must be maintained to prevent bleeding in factor-
deficient patients. Therapy for a hemophilic patient is designed
to maintain the factor level above 30%. Depending on the BOX 40-3 Other Plasma Procoagulants
patients condition, a higher level may be desirable, such as in Fibrinogen
a patient undergoing surgery. The half-life is also important in Factor XIII
monitoring anticoagulant therapy, especially warfarin (Cou- Phospholipids
madin), because factor VII is reduced rapidly (6 hours), whereas Calcium
reduction of prothrombin takes 4 to 5 days. Therefore, the full Von Willebrand factor
effect of warfarin is not realized until approximately 5 days
after therapy has begun.
Factor XIIIa is a transglutaminase that catalyzes the transfer of
Classification of Procoagulants amino acids among the chains of fibrin polymers. This reac-
Physiologic Function of Procoagulants tion cross-links fibrin polymers to provide physical strength to
The plasma procoagulants may be serine proteases or cofac- the fibrin clot. Factor XIIIa reacts with other plasma and cel-
tors.24 Serine proteases are proteolytic enzymes of the trypsin lular structural proteins and is essential to wound healing and
family and include the procoagulants thrombin (factor IIa); tissue integrity. Factor XIII is a heterodimer whose subunit
factors VIIa, IXa, Xa, XIa, and XIIa; and prekallikrein (pre-K).25 is produced mostly from megakaryocytes and monocytes. The
Each member has a reactive seryl amino acid residue in its subunit is produced in the liver.28
active site and acts on its substrate by hydrolyzing peptide Coagulation occurs on the surface of platelet or endothelial
bonds, digesting the primary backbone and producing small cell membrane phospholipids and not in fluid phase. Any fluid
polypeptide fragments. Serine proteases are synthesized as phase reaction is inhibited by the coagulation control proteins.
inactive zymogens consisting of a single peptide chain (Table Serine proteases bind to negatively charged phospholipid sur-
40-6). Activation occurs when the zymogen is cleaved at one faces, mostly phosphatidylserine, through positively charged
or more specific sites by the action of another protease during calcium ions, so calcium is required for the assembly of coagu-
the coagulation process. Activation is a localized cell-surface lation complexes on phospholipid membranes. VWF is a large
process, limited to the site of injury and controlled by regula- glycoprotein that participates in platelet adhesion and trans-
tory mechanisms. If zymogen activation is uncontrolled and ports the procoagulant factor VIII. VWF is synthesized in mega-
generalized, the condition is called disseminated intravascular karyocytes and endothelial cells.29
coagulation (DIC), a serious, often life-threatening condition
(see Chapter 42). Biochemical Nature of Several Procoagulants
The coagulation cofactors are TF, factor V, factor VIII, and Vitamin KDependent Prothrombin Group. Pro-
HMWK. Each cofactor binds its particular serine protease. thrombin, factors VII, IX, and X, and the regulatory proteins
When bound, serine proteases gain stability and increased reac- protein C, protein S, and protein Z are vitamin K dependent
tivity (Table 40-7).26 (Table 40-8). These are named the prothrombin group because of
The remaining components of the coagulation pathway are their structural resemblance to prothrombin: all seven proteins
fibrinogen, factor XIII, phospholipids, calcium, and VWF (Box have 10 to 12 glutamic acid units near their amino termini.
40-3). Fibrinogen is the ultimate substrate of the coagulation Vitamin K is a quinone found in green leafy vegetables, fish,
pathway. When hydrolyzed by thrombin, fibrinogen polymer- and liver, and is produced by the intestinal organisms Bacteroi-
izes to form the primary structural protein of the fibrin clot.27 des fragilis and Escherichia coli. Vitamin K catalyzes an essential
634 PART VIII Hemostasis and Thrombosis

Warfarin

Epoxide
reductase

VK hydroxyquinone VK epoxide
Ca2
COO OOC
COO

 CH2 Carboxylase  CH2

 CH2  CH2
O2 CO2
HN2 CH COOH HN2 CH COOH

Glu Gla

Ca2 NH2
Ca2
10 to 12 Gla Ca2
Ca2
Ca2 COOH
Prothrombin
Figure 40-8 Vitamin K (VK) posttranslational -carboxylation of coagulation factors II (prothrombin), VII, IX, and X, and control proteins C, S, and Z. Vitamin
K hydroxyquinone transfers a carboxyl (COO) group to the carbon of glutamic acid (Glu), creating -carboxyglutamic acid (Gla). The negatively charged
pocket formed by the two carboxyl groups attracts ionic calcium, which enables the molecule to bind to phosphatidylserine. Vitamin K hydroxyquinone is
oxidized to vitamin K epoxide by carboxylase in the process of transferring the carboxyl group but is subsequently reduced to the hydroxyquinone form by
epoxide reductase. Warfarin suppresses epoxide reductase, which slows the reaction and prevents -carboxylation. Des-carboxy proteins are unable to
participate in coagulation. There are typically 10 to 12 -carboxyglutamic acid molecules near the amino terminus of the vitamin Kdependent factors.

posttranslational modification of the prothrombin group pro- Platelet membrane Platelet membrane
teins: -carboxylation of amino-terminal glutamic acids (Figure binding site: GP Ib/V/IX binding site: GPIIb/IIIa
40-8). Glutamic acid is modified to -carboxyglutamic acid
when a second carboxyl group is added to the carbon. With
two ionized carboxyl groups, the -carboxyglutamic acids gain VWF VIII Factor VIII
0.620 MD 260 kD reactive site
a net negative charge, which enables them to bind ionic calcium
(Ca2+). The bound calcium permits the vitamin Kdependent
proteins to bind negatively charged phospholipids, such as
phosphatidylserine. Phospholipid binding is essential to coag- Collagen binding site
ulation reactions. Figure 40-9 Von Willebrand factor (VWF)factor VIII complex. Factor VIII
In vitamin K deficiency or in the presence of warfarin, a circulates covalently bound to VWF. VWF provides three other active receptor
vitamin K antagonist, the vitamin Kdependent procoagulants sites: VWF binds to collagen and binds to glycoprotein Ib/IX/V to support
are released from the liver without the second carboxyl group platelet (PLT) adhesion and binds to glycoprotein IIb/IIIa to facilitate PLT
added to the carbon. These are called des--carboxyl proteins or aggregation.
proteins in vitamin K antagonism (PIVKAs). Because they lack the
second carboxyl group, they cannot bind to Ca2+ and phospho- VWF has receptor sites for both platelets and collagen
lipid, so they cannot participate in the coagulation reaction. (Figure 40-9) and helps bind platelets to exposed subendothe-
Vitamin K antagonism is the basis for oral anticoagulant (war- lial collagen during platelet adhesion, especially in arteries and
farin) therapy (see Chapter 46). arterioles where the flow of blood is faster. The primary platelet
surface receptor for VWF is GP Ib/IX/V.31 Arginineglycine
Von Willebrand Factor: Factor VIII Complex. VWF is aspartic acid (RGD) sequences in VWF also bind a second
a multimeric glycoprotein composed of multiple subunits of platelet integrin, GP IIb/IIIa, during platelet aggregation.
240,000D each.30 The subunits are produced by endothelial A third site binds to collagen, and a fourth site binds the
cells and megakaryocytes, where they combine to form mole- plasma procoagulant cofactor, factor VIII. Factor VIII is pro-
cules that range from 600,000 to 20,000,000D. VWF mole- duced by hepatocytes and other tissues. It is one of two plasma
cules are stored in -granules in platelets and in storage sites procoagulants whose production is sex linked, the other being
called Weibel-Palade bodies in endothelial cells. The molecules factor IX. Factor VIII has a molecular mass of 260,000D and
are released from storage into the plasma, where they circulate circulates bound to VWF. Free factor VIII is unstable except
at a concentration of 7 to 10mcg/mL. during coagulation and cannot be detected in plasma. Factor
CHAPTER 40 Normal Hemostasis and Coagulation 635

Fibrinogen Structure
Trinodular structure

Enlarged showing chains

B A A B


D domain D domain
E domain

Supercoiled
region
A peptide
B peptide
Disulfide bonds
Figure 40-10 Structure of fibrinogen. Fibrinogen is a trinodular structure composed of three pairs (A, B, ) of disulfidebonded polypeptide chains. The
central node is known as the E domain. Thrombin cleaves small peptides, A and B, from the and chains in this region to form fibrin. The central nodule
is joined by supercoiled -helices to the terminal nodules known as the D domains. (From McKenzie SB, Williams JL: Clinical laboratory hematology, ed 2,
Upper Saddle River, NJ, 2009, Pearson, p 653.)

VIII is labile and deteriorates over hours in stored blood despite portions of the D domain of neighboring monomers, sponta-
being bound to VWF. Males with hemophilia A have dimin- neously polymerizing to form fibrin polymer (Figure 40-11).
ished factor VIII activity but normal VWF levels.32 Because Factor XIIIa catalyzes the formation of covalent bonds
factor VIII depends on VWF for stability, individuals with von between the carboxy termini of chains from adjacent D
Willebrand disease who have diminished VWF also have domains. These bonds link the -amino acid of lysine moieties
diminished factor VIII activity levels. Typically, factor VIII and the -amide group of glutamine units. Multiple cross-links
levels decrease to hemorrhagic levels (less than 30%) only in form to provide an insoluble meshwork of fibrin polymers,
severe von Willebrand disease. linked by their D domains. Cross-linking also covalently incor-
porates fibronectin, a plasma protein involved in cell adhesion,
Fibrinogen Structure and Fibrin Formation. Fibrino- and 2-antiplasmin, rendering the fibrin mesh resistant to fibri-
gen is the primary substrate of thrombin. It is a 340,000-D nolysis. Plasminogen, the primary serine protease of the fibri-
glycoprotein synthesized in the liver. The normal plasma nolytic system, also becomes covalently bound via lysine
concentration of fibrinogen ranges from 200 to 400mg/dL, moieties, as does TPA, a serine protease that ultimately hydro-
the highest concentration of all the plasma procoagulants. lyzes and activates bound plasminogen to initiate fibrinolysis.
Platelet -granules absorb, transport, and release abundant
fibrinogen.33 Coagulation Pathways
The fibrinogen molecule is a mirror-image dimer, with each Tissue Factor Pathway
half consisting of three nonidentical polypeptides, designated TF, a membrane receptor for factor VIIa, is present on vascular
, , and , united by disulfide bonds (Figure 40-10). The six cells that are not normally in contact with blood, such as fibro-
amino termini assemble to form a bulky central region called blasts. Coagulation is triggered by the exposure of TF to plasma
the E domain. The carboxy termini assemble at the two ends of proteins on injury.35 In inflammatory conditions, leukocytes
the molecule to form two D domains.34 and other cells can also express TF and initiate coagulation.
Thrombin cleaves fibrinopeptides A and B from the protrud- Factor VIIa binds to TF in the presence of phospholipid and
ing amino termini of each of the two and chains, reducing calcium and triggers the activation of both factor IX and factor
the overall molecular weight by 10,000D. The cleaved fibrino- X (Figures 40-1 and 40-12). Factor IXa binds factor VIIIa on
gen is called fibrin monomer. The exposed fibrin monomer platelet phospholipid surfaces; this complex also activates
and chain ends (E domain) have an immediate affinity for factor X.
636 PART VIII Hemostasis and Thrombosis

A A
B B

Fibrinogen D E D
Thrombin cleaves
fibrinopeptides A and B
Fibrin monomer D E D

Spontaneous polymerization

D E D
Fibrin polymer
D E D D E D
XIIIa cross-links D domains
Stabilized fibrin D E D D E D
D E D D E D D E D
D E D D E D

Figure 40-11 Formation of a stabilized fibrin clot. Thrombin cleaves fibrinopeptides A and B to form fibrin monomer. Fibrin monomers polymerize due to
the affinity of thrombin-cleaved positively charged E domains for negatively charged D domains of other monomers. Factor XIIIa catalyzes the covalent cross-
linking of chains of adjacent D domains to form a urea-insoluble stable fibrin clot. (Modified from McKenzie SB, Williams JL: Clinical laboratory hematology,
ed 2, Upper Saddle River, NJ, 2009, Pearson, p 654.)

Initiation: vascular
wall injury exposes
fibroblast (FB)
tissue factor (TF) P

FB IX
2 TF
Ca a
P VII
XIa XI
VII
VIII
IXa VIIIa
Ca2 P Amplification and
propagation: activated
X Xa Va V platelet (P) surfaces
2 P
Ca
XIII

Prothrombin Thrombin
d P
XIIIa ke
l- in lot P
s c
os in P
Fibrinogen Fibrin Cr fibr P
P
P
P

Figure 40-12 Coagulation pathway. Initiation occurs on the membrane of extravascular cells when tissue factor (TF) is exposed to blood through injury or
inflammation. Factor VIIa binds to TF and activates both factor IX and factor X. This is also known as the extrinsic pathway. Amplification and propagation
occur on the surface of activated platelets. Factor IXa, with its cofactor VIIIa, activates factor X. Factor Xa, with its cofactor Va, converts prothrombin to thrombin
(common pathway). Thrombin cleaves fibrinogen to fibrin and also activates factors V, VIII, XI, and XIII. Factor XIa activates factor IX, which further activates
factor X and generates thrombin. This is the intrinsic pathway. (Modified from Marques MB, Fritsma GA: Quick guide to coagulation, ed 2, Washington, DC,
2009, American Association for Clinical Chemistry Press.)

Factor Xa, activated by both TF:VIIa and IXa:VIIIa, binds also activates factor XIII, which cross-links and stabilizes fibrin
factor Va on phospholipid surfaces. This Xa:Va complex acti- (see Figure 40-11).
vates prothrombin in a multistep hydrolytic process that The tissue factor pathway is characterized by three multimo-
releases a peptide fragment from prothrombin called prothrom- lecular complex formations. The first complex is composed of
bin 1+2. Activated prothrombin is termed thrombin. Thrombin TF, factor VIIa, phospholipid, and Ca2+ on the TF-bearing cell;
cleaves fibrinopeptides A and B from plasma fibrinogen; this this complex activates IX and X. The second is composed of
produces fibrin monomer, which then polymerizes. Thrombin factor IXa, factor VIIIa, phospholipid, and Ca2+ on platelets,
CHAPTER 40 Normal Hemostasis and Coagulation 637

TABLE 40-9 Coagulation Cascade Complexes pathway leading to thrombin generation. Most coagulation
experts identified the activation of factor XII as the primary step
Components Function
in coagulation because this factor could be found in blood,
First complex VIIa, tissue factor, Cleaves IX and X whereas TF could not. Consequently, the reaction system that
phospholipid, and Ca2+ begins with factor XII and culminates in fibrin polymerization
Second complex IXa, VIIIa, phospholipid, Cleaves X, called tenase has been called the intrinsic pathway. The coagulation factors of
and Ca2+ the intrinsic pathway, in order of reaction, are XII, pre-K,
Third complex Xa, Va, phospholipid, Cleaves prothrombin, HMWK, XI, IX, VIII, X, V, prothrombin, and fibrinogen (see
and Ca2+ called prothrombinase Figure 40-1). The laboratory test that screens for these factors
is the activated partial thromboplastin time (APTT or PTT). We
now know that the contact factors XII, pre-K, and HMWK do
sometimes called tenase; this complex also activates factor X. not play a significant role in in vivo coagulation, although their
The third complex is composed of factor Xa, factor Va, phos- deficiencies cause abnormal results in in vitro laboratory tests
pholipid, and Ca2+ and is often called prothrombinase; this of the intrinsic pathway, such as the PTT.
complex converts prothrombin to thrombin (Table 40-9). Because TF is not present in blood, the tissue factor pathway
has been called the extrinsic pathway. This pathway includes the
Contact Factors factors VII, X, V, prothrombin, and fibrinogen. Initiation of
The contact factor complex, composed of factor XIIa, HMWK coagulation by TF:VIIa is the principal mechanism in vivo for
(Fitzgerald), and pre-K (Fletcher), activates factor XI. Factor generating thrombin. The test used to measure the integrity of
XIIa transforms pre-K, a glycoprotein that circulates bound to the extrinsic pathway is the prothrombin time test (PT). See
HMWK, into its active form kallikrein, which cleaves HMWK Chapter 46 for discussion of PT and PTT, two important screen-
to form bradykinin. Deficiencies of factor XII, HMWK, or pre-K ing tests. Factor IX and factor VIII are not included in the
do not cause clinical bleeding disorders. However, deficiencies extrinsic pathway, because they are not measured by the PT test.
do produce prolonged results on laboratory screening tests and But clearly, the IXa:VIIIa complex is crucial to the activation of
necessitate investigation. Factor XII is activated in vitro by factor X. Deficiencies of either one of these components of the
contact with negatively charged surfaces, such as nonsilicon- tenase complexfactor VIII in hemophilia A, or factor IX in
ized glass or the ellagic acid in the partial thromboplastin time hemophilia Bcan result in severe and life-threatening hemor-
(PTT) reagent. In vivo, foreign materials such as stents or valve rhage. The two pathways have in common factor X, factor V,
prostheses may activate contact factors to cause thrombosis. prothrombin, and fibrinogen; this portion of the coagulation
pathway is often called the common pathway. These designations
Factor XI Activation Pathway intrinsic, extrinsic, and commonare used extensively to inter-
Factor XI is activated by thrombin, as well as by the contact pret in vitro laboratory testing and to identify factor deficiencies;
factors. Factor XIa activates factor IX, and the reaction proceeds however, they do not adequately describe the complex interde-
as described previously. Deficiencies of factor XI (Rosenthal pendent reactions in the in vivo coagulation process.
syndrome) usually result in mild bleeding disorders.36
Cell-Based (In vivo) Coagulation
Thrombin A spectacular combination of cellular and biochemical events
The primary function of thrombin is to cleave fibrinopeptides function in harmony to keep blood liquid within the veins and
A and B from the and chains of the fibrinogen molecule, arteries, prevent blood loss from injuries by the formation of
triggering fibrin polymerization (see Figure 40-11). In addi- thrombi, and reestablish blood flow during the healing
tion, thrombin amplifies the coagulation mechanism by acti- process.37 As noted earlier, the series of cascading proteolytic
vating cofactors V and VIII and factor XI. Thrombin also reactions traditionally known as the extrinsic and intrinsic coag-
activates factor XIII, the fibrin-stabilizing factor. Factor XIIIa ulation pathways do not fully describe how coagulation occurs
forms covalent bonds between the D domains of the fibrin in vivo. These pathways are not distinct independent alterna-
polymer to cross-link and stabilize the fibrin clot. Thrombin tive mechanisms for generating thrombin, but are actually very
also initiates aggregation of platelets. Thrombin bound to interdependent. For example, a deficiency of factor VII in the
thrombomodulin activates the protein C pathway to suppress extrinsic pathway can cause significant bleeding, even when the
coagulation, and it activates TAFI to suppress fibrinolysis. intrinsic pathway is normal. Similarly, deficiencies of some
Thrombin therefore plays a role in coagulation (fibrin), in intrinsic pathway proteins, notably factors VIII and IX, may
platelet activation, in coagulation control (protein C), and in cause severe bleeding problems, regardless of the presence of
fibrinolysis (TAFI). Because of its multiple autocatalytic func- a normal extrinsic pathway.38
tions, thrombin is considered the chief protease of the coagula- Current thinking about the physiologic process of coagula-
tion pathway. tion recognizes the contribution of specific cells in regulating
hemostasis. The cell-based model of hemostasis (Figure 40-13)
Extrinsic, Intrinsic, and Common Pathways describes the interaction of two cell types: platelets, within the
Before 1992, two alternative coagulation pathways were blood circulation, and extravascular cells that bear TF.39 The
described, both of which activated factor X in a common cell-based model of coagulation is often described as occurring
638 PART VIII Hemostasis and Thrombosis

Small amount VIII VWF


VIIa of thrombin

Thr
TF VIIa Xa Va VIIIa

Tissue factor-
bearing cell Platelet
XIa
Va
TF VIIa

Xa

IXa VIIIa Va

Thrombin
Activated COAT
platelet
burst
Initiation
Amplification
Propagation

Figure 40-13 In vivo cell-based coagulation. Factor VIIa binds to tissue factor (TF) and activates both X and IX. Cell-bound factor Xa combines with Va
and generates a small amount of thrombin, which activates platelets and factors V, VIII, and XI. Factor IXa, activated by both TF:VIIa and XIa, combines with
VIII on the platelet surface to activate X, which forms prothrombinase (Xa:Va), and produces a burst of thrombin that cleaves fibrinogen into fibrin.

in three separate but overlapping phases: initiation, amplifica- ready to go. Platelets are activated at the site of injury by both
tion, and propagation. the low-level thrombin generated in the initiation phase and
by adherence to exposed collagen. They are sometimes referred
Initiation to as COAT platelets, platelets partially activated by collagen and
Continuous low-level activation of the TF pathway occurs on thrombin.40 These partially activated COAT platelets have a
TF-bearing cells outside the circulation, even in the absence of higher level of procoagulant activity than platelets exposed to
injury. Fibroblasts and other subendothelial cells carry TF, a collagen alone. Thrombin, in addition to activating platelets,
transmembrane protein that is a cofactor to factor VII. Smaller activates factor V released from platelet -granules, activates
plasma coagulation proteins, such as factors VII, IX, and X, are factor VIII and dissociates it from VWF, and activates factor XI.
present in extravascular fluid as well as in the circulation.
About 1% or less of VII is present normally in the activated Propagation
form.20 VIIa binds to TF, and the TF:VIIa complex activates In the propagation phase the reactions occur on the surface of
small amounts of both factor IX and factor X. Factor Xa com- the activated platelet, which now has all the components
plexes with Va to form prothrombinase, Xa:Va, and generates needed for coagulation. Large numbers of platelets adhere to
a limited amount of thrombin. Factor Va comes from the acti- the site of injury. Factor IXa from the initiation phase binds to
vation of factor V by Xa, or by platelets if there has been an VIIIa on the platelet. More IXa is generated from platelet-
injury, or by noncoagulation proteases.40 Xa:Va bound to the bound XIa. The IXa:VIIIa complex (tenase) activates Xa, which
cell is protected from inactivation by control proteins. However, complexes with Va to activate prothrombin and generate a
if Xa:Va dissociates from the cell, it is rapidly inactivated by burst of thrombin, which cleaves fibrinogen into a fibrin clot.
the protease inhibitors TFPI, antithrombin, and protein Thrombin also activates factor XIII to stabilize the clot, binds
Zdependent protease inhibitor (ZPI). Factor IXa, on the other to thrombomodulin to activate the protein C control pathway,
hand, can move to other cells through the extravascular fluid, and activates TAFI to inhibit fibrinolysis.
because it is inhibited more slowly by antithrombin and is not Because coagulation is dependent on the presence of both
inhibited by TFPI or ZPI. Because the larger components of TF-bearing cells and activated platelets, clotting is localized to
hemostasisplatelets and VIII:VWFare not present in the the site of injury. Protease inhibiters and intact endothelium
extravascular fluid, factor IX activation does not proceed further prevent clotting from spreading to other parts of the body.
until there is an injury. The low level of thrombin generated In the cell-based model, it may be helpful to think of the
by the cell-bound Xa:Va in the initiation phase does not extrinsic or TF pathway as occurring on the TF-bearing cell and
produce a fibrin clot. the intrinsic pathway (minus factor XII, HMWK, and pre-K) as
occurring on the platelet surface. However, these are not totally
Amplification separate pathways. TF:VIIa activates both factor IX and factor
When an injury and bleeding occur, platelets and VIII:VWF X. Factor XI is activated by the thrombin produced in the initia-
spill into the extravascular space. Coagulation is primed and tion phase, bypassing factor XII and the contact factors of the
CHAPTER 40 Normal Hemostasis and Coagulation 639

intrinsic pathway. Both pathways are necessary for normal Acquired or inherited deficiencies of these proteins may be
coagulation, and deficiencies of VII, IX, VIII, X, V, or II can associated with increased incidence of venous thromboem-
cause serious bleeding problems. Factor XI functions to supple- bolic disease. Figure 40-14 illustrates coagulation mechanism
ment or boost factor IX activation, so deficiencies of factor XI regulatory points. Characteristics of these and other coagula-
usually produce less severe clinical manifestations than defi- tion regulatory proteins are summarized in Table 40-10.
ciencies of the other factors.36 The contact factors XII, HMWK,
and pre-K are not involved in physiologic hemostasis, so defi- Tissue Factor Pathway Inhibitor
ciencies of these factors do not cause bleeding, although they Factor VIIa and TF combine to activate factors IX and X in the
do prolong the PTT. tissue factor pathway. Factor Xa reacts with TF:VIIa and binds
TFPI, an important coagulation regulatory protein (Figure
40-15). TFPI is synthesized by endothelial cells.41 About 80%
COAGULATION REGULATORY MECHANISMS
is bound to the vessel wall and 20% circulates in the plasma.
Serine proteases and cofactors in the coagulation system are Most of the plasma TFPI is truncated and bound to low-density
regulated by inhibitors, cofactors, and feedback loops to main- lipoproteins. The full-length free form, comprising only 2% of
tain a complex and delicate balance between thrombosis the total TFPI, is an effective inhibitor of the tissue factor
and abnormal bleeding. The principal regulators are TFPI, pathway. TFPI inhibits coagulation in a two-step process by
antithrombin, and the protein C pathway. These function as inactivating Xa and then binding to TF:VIIa to prevent it from
natural anticoagulants, because they inhibit the action of pro- activating additional Xa.42,43 TFPI provides feedback inhibition,
coagulants and prevent excessive clot formation or thrombosis. because it is not functional until X is activated. Protein S, the

AT XIa ZPI
VIIa
TFPI
TF

AT IXa VIIIa APC

AT

TFPI Xa Va APC
ZPI

AT Thr XIIIa

Cross-linked
Fibrinogen Fibrin polymer fibrin

Figure 40-14 Coagulation pathway, showing regulatory points. APC, Activated protein C; AT, antithrombin; TFPI, tissue factor pathway inhibitor; ZPI, protein
Zdependent protease inhibitor.

TABLE 40-10 Coagulation Regulatory Proteins


Molecular Mass Half-Life Mean Plasma
Name Function (daltons) (hours) Concentration
Tissue factor pathway inhibitor With Xa, binds tissue factor:VIIa 33,000 Unknown 60-80ng/mL
Thrombomodulin Endothelial cell surface receptor for thrombin 450,000 Does not circulate None
Protein C Serine protease 62,000 7-9 2-6mcg/mL
Protein S Cofactor 75,000 Unknown 20-25mcg/mL
Antithrombin Serpin 58,000 68 24-40mg/dL
Heparin cofactor II Serpin 65,000 60 30-70mcg/mL
Z-dependent protease inhibitor Serpin 72,000 Unknown 1.5mcg/mL
1-Protease inhibitor (1-antitrypsin) Serpin 60,000 Unknown 250mg/dL
2-Macroglobulin Serpin 725,000 60 150-400mg/dL

Serpin, Serine protease inhibitor.


640 PART VIII Hemostasis and Thrombosis

X Xa TFPI

VIIa VIIa Xa
Ca2+ TF Ca2+ Ca2+ TF Ca2+

Subendothelial cell membrane Subendothelial cell membrane

Figure 40-15 Tissue factor pathway inhibitor (TFPI) binds the complex of tissue factor (TF), factor VIIa, and factor Xa. TFPI inactivates both Xa and the
TF:VIIa complex by a feedback mechanism that requires activation of Xa.

Endothelial hydrolyzes and inactivates factors Va and VIIIa, slowing or


cell membrane blocking coagulation.
Protein S, the cofactor that binds and stabilizes APC, is
TM Thrombin synthesized in the liver and circulates in the plasma in two
forms: free and covalently bound to the complement control
Endothelial cell or protein C4b-binding protein (C4bBP). Bound protein S cannot
Va VIIIa
platelet membrane participate in the protein C anticoagulant pathway; only free
plasma protein S can serve as the APC cofactor. Protein
PC APC PS
SC4bBP binding is of particular interest in inflammatory con-
ditions, because C4bBP is an acute phase reactant. When the
plasma C4bBP level increases, additional protein S is bound,
and free protein S levels become proportionally decreased,
Free PS 40% Vi VIIIi
which increases the risk of thrombosis. Chronic acquired or
inherited protein C or protein S deficiency, or mutations of
protein C, protein S, or factor V, compromise downregulation
of Va and VIIIa and may be associated with recurrent venous
PS Bound PS 60%
thromboembolic disease, which underscores the importance of
the protein C regulatory system (see Chapter 42).
C4bBP

Figure 40-16 Protein C pathway. After binding thrombomodulin (TM), Serine Protease Inhibitors (Serpins)
thrombin activates protein C (PC). Free protein S (PS) binds and stabilizes Antithrombin was the first of the coagulation regulatory pro-
activated protein C (APC). The activated protein Cprotein S complex digests teins to be identified and the first to be assayed routinely in
and inactivates factors Va and VIIIa. C4bBP, C4b binding protein; Vi, inacti- the clinical hemostasis laboratory.47 Other members of the
vated factor V; VIIIi, inactivated factor VIII. serpin family are heparin cofactor II, ZPI, protein C inhibitor,
1-protease inhibitor (1-antitrypsin), 2-macroglobulin, 2-
antiplasmin, and PAI-1.48
cofactor of activated protein C (APC), is also a cofactor of TFPI Antithrombin is a serine protease inhibitor (serpin) that
and stimulates Xa inhibition by TFPI.44 Because of the inhibi- binds and neutralizes all serine proteases except factor VIIa,
tory action of TFPI, the TF:VIIa:Xa reaction is short-lived. including thrombin (factor IIa) and factors IXa, Xa, XIa, XIIa,
Coagulation pathway amplification occurs primarily through kallikrein, and plasmin. Heparin cofactor II is a serpin that
IXa,45 because Xa activation by IXa:VIIIa is negligibly affected inactivates primarily thrombin. Antithrombin and heparin
by TFPI. cofactor II both require heparin for effective anticoagulant
activity. In vivo, heparin is available from endothelial associ-
Protein C Regulatory System ated mast cell granules or as endothelial cell heparan sulfate,
During thrombosis, thrombin propagates the clot as it a natural glycosaminoglycan that activates antithrombin,
cleaves fibrinogen and activates factors V, VIII, XI, and XIII. although not to the same intensity as unfractionated heparin.
In intact normal vessels, where coagulation would be inex Therapeutically, unfractionated heparin, low-molecular-weight
pedient, thrombin binds the endothelial cell membrane heparin, and heparin pentasaccharide are administered as
protein thrombomodulin and triggers an essential coagulation anticoagulants. Unfractionated heparin is a heterogeneous
regulatory system called the protein C system.46 The thrombin- glycosaminoglycan composed of 15 to 30 saccharide units.
thrombomodulin complex activates the zymogen protein C to Unfractionated heparin increases the ability of antithrombin
a serine protease (Figure 40-16). APC binds its cofactor, free to neutralize thrombin and factors IXa, Xa, XIa, and XIIa by
plasma protein S. The stabilized APCprotein S complex 1000-fold. Any heparin molecule of 17 or more saccharide
CHAPTER 40 Normal Hemostasis and Coagulation 641

Protease Active
binding protease
site site

AT Thr

Heparin Heparin
binding site binding site
AT Thr

Heparin with
17 sugar units

AT Thr

Figure 40-17 Unfractionated heparin potentiates the reaction between antithrombin (AT) and thrombin (Thr, factor IIa). AT attaches to a specific pentasac-
charide sequence in unfractionated heparin. The thrombin binding site for heparin is adjacent to the AT site (approximation). The AT is sterically modified
(allostery) to covalently bind and inactivate the thrombin active protease site. Thrombin and AT, covalently bound, release from heparin and form measurable
plasma thrombin-antithrombin (TAT) complexes, useful as a marker of coagulation activation.

units simultaneously binds antithrombin and thrombin. The thrombin-thrombomodulin complex, TPA, and urokinase.48 It
antithrombin covalently binds and inactivates a thrombin mol- is found not only in plasma but in many other body fluids
ecule, forming an inactive thrombin-antithrombin complex, and organs. Depending on its target, it can function as an
which is released from the heparin molecule. Laboratory mea- anticoagulant (inhibits thrombin), as a procoagulant (inhibits
surement of thrombin-antithrombin is used as an indicator for thrombin-thrombomodulin and APC), or as a fibrinolytic
thrombosis. Unfractionated heparins catalytic effect is to inhibitor (inhibits TPA).
approximate thrombin to antithrombin (i.e., to bring the two The serpins 1-protease inhibitor and 2-macroglobulin are
molecules into proximity with each other) and to induce allo- able to inhibit serine proteases reversibly. See Table 40-11 and
stery, or change in steric conformation, of the antithrombin the section on fibrinolysis for further information on 2-
molecule (Figure 40-17). Heparin fractions of less than 17 antiplasmin and PAI-1.
saccharide units, such as therapeutic low-molecular-weight
heparin, are able to bind antithrombin or thrombin separately
FIBRINOLYSIS
but cannot bring them into the necessary spatial configuration
to bind each other. Nevertheless, the allosteric changes Fibrinolysis, the final stage of coagulation (Figure 40-18),
induced in the antithrombin still make it capable of binding begins a few hours after fibrin polymerization and cross-
and inactivating factor Xa and other serine proteases. To sum- linking. Plasminogen, plasmin, TPA, and PAI-1, discussed
marize, with unfractionated heparin therapy, antithrombin later, are incorporated into the fibrin clot by binding to lysine
preferentially inactivates thrombin, and with low-molecular- through kringle loops. Fibrinolysis is the systematic, acceler-
weight heparin therapy, antithrombin preferentially inactivates ating hydrolysis of fibrin by bound plasmin, which cleaves
factor Xa. peptide bonds at arginine and lysine moieties in regions con-
ZPI, in the presence of its cofactor, protein Z, is a potent necting fibrins D and E domains (Figure 40-19).52 TPA and
inhibitor of factor Xa.49-51 ZPI covalently binds protein Z and urokinase activate fibrin-bound plasminogen several hours
Xa in a complex with Ca2+ and phospholipid. Protein Z is a after thrombus formation, degrading fibrin and restoring
vitamin Kdependent plasma glycoprotein that is synthesized normal blood flow during vascular repair. Again, there is a
in the liver. Although protein Z has a structure similar to that delicate balance between activators and inhibitors. Excessive
of other vitamin Kdependent proteins VII, IX, X, and protein fibrinolysis can lead to bleeding due to premature clot lysis
C, it lacks an activation site and, like protein S, is nonproteo- before wound healing is established, whereas inadequate fibri-
lytic. Protein Z increases the ability of ZPI to inhibit Xa 1000- nolysis can lead to clot extension and thrombosis.
fold. ZPI also inhibits factor XIa, in a separate reaction that
does not require protein Z, phospholipid, and Ca2+. The Plasminogen and Plasmin
inhibition of XIa is accelerated twofold by the presence of Plasminogen is a 92,000-D plasma zymogen produced by
heparin. the liver (see Table 40-11).53,54 It is a single-chain protein
Protein C inhibitor is a nonspecific, heparin-binding serpin possessing five glycosylated loops termed kringles. Kringles
that inhibits a variety of proteases, including APC, thrombin, enable plasminogen to bind fibrin lysine residues during
642 PART VIII Hemostasis and Thrombosis

TABLE 40-11 Proteins of the Fibrinolysis Pathway


Molecular Mass Mean Plasma
Name Function (daltons) Half-Life Concentration
Plasminogen Plasma serine protease, plasmin digests 90,000 24-26hr 15-21mg/dL
fibrin/fibrinogen
Tissue plasminogen activator Serine protease secreted by activated endothelium, 68,000 Unknown 4-7mcg/dL
activates plasminogen
Urokinase Serine protease secreted by kidney, activates 54,000 Unknown
plasminogen
Plasminogen activator inhibitor 1 Secreted by endothelium, inhibits tissue 52,000 1hr 14-28mg/dL
plasminogen activator
2-Antiplasmin Inhibits plasmin 51,000 Unknown 7mg/dL
Thrombin-activatable fibrinolysis inhibitor Suppresses fibrinolysis 55,000 8-10min 5mcg/mL

Fibrinogen Fibrin FDPs, D-dimer

Bound
TPA plasmin TAFI

Plasminogen

TPA PAI-1

Free
plasmin

2-
antiplasmin

Figure 40-18 Fibrinolysis pathway and inhibitors. Plasminogen and tissue plasminogen activator (TPA) are bound to fibrin during coagulation. TPA converts
bound plasminogen to plasmin, which slowly digests fibrin to form fibrin degradation products (FDPs) X, Y, D, E, and D-D (D-dimer). D-dimer is produced from
cross-linked fibrin. Free plasmin is neutralized by 2-antiplasmin. TPA is neutralized by plasminogen activator inhibitor 1 (PAI-1). Thrombin-activatable fibri-
nolysis inhibitor (TAFI) inhibits fibrinolysis by binding to lysine residues on fibrin, preventing the binding of plasminogen and TPA.

polymerization. This binding is essential to fibrinolysis. Plas- Plasminogen Activation and Control
minogen likewise binds plasma 2-antiplasmin, the major Tissue Plasminogen Activator
fibrinolysis control protein. Fibrin-bound plasminogen Endothelial cells secrete TPA, which hydrolyzes fibrin-bound
becomes converted into a two-chain active plasmin molecule plasminogen and initiates fibrinolysis. TPA, with two glycosyl-
when cleaved between arginine at position 561 and valine ated kringle regions, forms covalent lysine bonds with fibrin
at position 562 by neighboring bound TPA or urokinase. during polymerization and segregates at the surface of the
Plasmin is a serine protease that systematically digests fibrin thrombus with plasminogen, where it begins the digestion
polymer by hydrolysis of arginine-related and lysine-related process by converting plasminogen to plasmin. Free TPA circu-
peptide bonds. As fibrin becomes digested, the exposed lates bound to inhibitors such as PAI-1 and is cleared from
carboxy-terminal lysine residues bind additional plasmin, plasma. Synthetic recombinant TPAs mimic intrinsic TPA and
which incrementally accelerates clot digestion. Plasmin is are a family of drugs used to dissolve pathologic clots that form
capable of digesting fluid-phase fibrinogen and factors V and in venous and arterial thrombotic disease.
VIII; however, localization to fibrin through lysine binding
prevents systemic activity. Plasma 2-antiplasmin rapidly binds Urokinase
and inactivates free plasmin. Several aminocarboxalic acids Urinary tract epithelial cells, monocytes, and macrophages
have an affinity for plasminogens kringles, which accounts for secrete another intrinsic plasminogen activator called uroki-
the antifibrinolytic properties of therapeutic tranexamic acid nase. Urokinase circulates in plasma at a concentration of
and -aminocaproic acid. 2 to 4ng/mL and becomes incorporated into the mix of
CHAPTER 40 Normal Hemostasis and Coagulation 643

Fibrinogen Fibrin

D E D D E D
D E D
E
D E D D D D E D
Plasmin D E D D E D

Plasmin
Fragment X D E D

Plasmin E D D
E
D D E D D
Fragment Y
D
E D
YY and
DXD
Plasmin complex

Plasmin

D D D
Fragments E DED complex,
D and E E D E E, and D-dimer

D D
Figure 40-19 Degradation of fibrinogen and fibrin by plasmin. Plasmin systematically degrades fibrinogen and fibrin by digestion of small peptides and
cleavage of DE domains. Fragment X consists of a central E domain with two D domains (D-E-D); further cleavage produces fragment Y (D-E), with eventual
degradation to D and E domains. From cross-linked fibrin, plasmin digestion produces fragment complexes from one or more monomers. D-dimer consists
of two D domains from adjacent monomers that have been cross-linked by XIIIa in the process of fibrin formation (thrombosis).

fibrin-bound plasminogen and TPA at the time of thrombus plasma protein that rapidly and irreversibly binds free plasmin.
formation. Urokinase has only one kringle region, does not 2-Antiplasmin also becomes cross-linked through lysine
bind firmly to fibrin, and has a relatively minor physiologic bonds to fibrin kringles during polymerization. There it slows
effect. Like TPA, purified urokinase preparations are used to fibrin digestion by plasmin.
dissolve thrombi in myocardial infarction, stroke, and deep
vein thrombosis. Thrombin-Activatable Fibrinolysis Inhibitor
TAFI is a plasma procarboxypeptidase that becomes activated
Plasminogen Activator Inhibitor 1 by the thrombin-thrombomodulin complex. This is the same
TPA and urokinase are prevented from activating free, fluid- complex that activates the protein C pathway; however, the
phase plasminogen by a plasma inhibitor, PAI-1. Probable two functions are independent. Activated TAFI inhibits fibri-
sources of PAI-1 include the endothelium, adipocytes, mega- nolysis by cleaving exposed carboxy-terminal lysine residues
karyocytes, and hepatocytes. Platelets store a pool of PAI-1, from partially degraded fibrin, preventing the binding of TPA
accounting for more than half of its availability and for its and plasminogen and blocking the formation of plasmin. In
delivery to the fibrin clot. PAI-1 is present in excess of the TPA coagulation factordeficient states, decreased thrombin pro-
concentration, and circulating TPA normally is bound to PAI-1. duction may reduce the activation of TAFI, resulting in
Only at times of endothelial cell activation, such as after increased fibrinolysis that contributes to bleeding. Conversely,
trauma, does the level of TPA secretion exceed that of PAI-1 to in thrombotic disorders, increased thrombin generation may
initiate fibrinolysis. Plasma PAI-1 levels vary widely. PAI-1 defi- increase the activation of TAFI. The resulting decreased fibri-
ciency has been associated with chronic mild bleeding due to nolysis may contribute further to thrombosis. TAFI also may
increased fibrinolysis; excess PAI-1 may be associated with play a role in regulating inflammation and wound healing.55
thrombosis due to reduction in fibrinolysis.
Fibrin Degradation Products and D-Dimer
2-Antiplasmin Fibrinolysis produces a series of identifiable fibrin fragments,
2-Antiplasmin is synthesized in the liver and is the primary X, Y, D, E, and D-D (see Figure 40-19).56 Several of these frag-
inhibitor of plasmin. Bound plasmin digests clots and restores ments inhibit hemostasis and contribute to hemorrhage by
blood vessel patency. Free plasmin in the circulation digests preventing platelet activation and by hindering fibrin polym-
plasma fibrinogen, factor V, factor VIII, and fibronectin, causing erization. Fragment X is described as the central E domain with
a potentially fatal primary fibrinolysis. 2-Antiplasmin is a the two D domains (D-E-D), minus some peptides cleaved by
644 PART VIII Hemostasis and Thrombosis

plasmin. Fragment Y is the E domain after cleavage of one D The various fragments may be detected by quantitative or
domain (D-E). Eventually these fragments are further digested semiquantitative immunoassay to reveal fibrinolytic activity.
to individual D and E domains. The D-D fragment, called D-dimer is separately detectable by a quantitative monoclonal
D-dimer, is two D domains from separate fibrin molecules antibody immunoassay. The D-dimer immunoassay is used to
cross-linked by the action of factor XIIIa. Fragments X, Y, D, identify chronic and acute DIC and to rule out venous throm-
and E are produced by digestion of either fibrin or fibrinogen boembolism in suspected cases of deep venous thrombosis or
by plasmin, but D-dimer is a product of digestion of fibrin. pulmonary embolism.

SUMMARY
The vascular intima, platelets, tissue factorbearing cells, and coag- Activation of coagulation pathways produces thrombin, which con-
ulation and fibrinolytic proteins interact to maintain hemostasis. verts fibrinogen to a fibrin clot. Thrombin also activates platelets
Intact vascular intima prevents coagulation through synthesis of and factors V, VIII, XI, and XIII, and binds to thrombomodulin to
prostacyclin, nitric oxide, TFPI, thrombomodulin, and heparan activate protein C and TAFI.
sulfate. Fibrinogen is cleaved by thrombin to form first fibrin monomer,
Damaged intima promotes coagulation by vasoconstriction, expo- then fibrin polymer, and finally, when acted on by factor XIIIa,
sure of TF and collagen, and secretion of VWF and other adhesion cross-linked fibrin.
molecules. In vivo, coagulation is initiated on TF-bearing cells. TF:VIIa acti-
Platelets function in primary and secondary hemostasis through vates factors IX and X, generating enough thrombin to activate
adhesion, aggregation, and secretion of granular contents. platelets and factors V, VIII, and XI. Coagulation proceeds on acti-
Platelets adhere to collagen through VWF and use fibrinogen to vated platelet phospholipid membranes with the formation of
aggregate. IXa:VIIIa and Xa:Va complexes, which produces a burst of throm-
Most coagulation factors are produced in the liver. bin that cleaves fibrinogen to fibrin.
The plasma factors of the prothrombin group (prothrombin; factors The coagulation pathway is regulated by TFPI, APC, and the
VII, IX, and X; protein C; protein S; and protein Z) require vitamin serpins, including antithrombin and ZPI. These control proteins
K in their production. prevent thrombosis and confine clotting to the site of injury.
Plasma coagulation factors include trypsinlike enzymes called The fibrinolytic pathway digests the thrombus. Plasminogen is
serine proteases and cofactors that stabilize the proteases. Factor converted to plasmin by TPA. Plasmin degrades fibrin to fragments
XIIIa is a transamidase. X, Y, D, and E, and D-dimer. Control proteins are PAI-1, 2-
The extrinsic pathway of coagulation consists of the membrane antiplasmin, and TAFI.
receptor TF and coagulation factors VII, X, V, II, and I. The PT is a
screening test for these factors. Now that you have completed this chapter, go back and
The intrinsic pathway factors are XII, pre-K, HMWK, XI, IX, VIII, X, read again the case study at the beginning and respond
V, II, and I. The PTT is a screening test for these factors. to the questions presented.

R E V I E W Q UESTIONS
1. What intimal cell synthesizes and stores VWF? 4. What role does vitamin K play for the prothrombin group
a. Smooth muscle cell factors?
b. Endothelial cell a. Provides a surface on which the proteolytic reactions
c. Fibroblast of the factors occur
d. Platelet b. Protects them from inappropriate activation by
compounds such as thrombin
2. What subendothelial structural protein triggers coagula- c. Accelerates the binding of the serine proteases and
tion through activation of factor VII? their cofactors
a. Thrombomodulin d. Carboxylates the factors to allow calcium binding
b. Nitric oxide
c. TF 5. What is the source of fibrinopeptides A and B?
d. Thrombin a. Plasmin proteolysis of fibrin
b. Thrombin proteolysis of fibrinogen
3. What coagulation plasma protein should be assayed when c. Proteolysis of prothrombin by factor Xa
platelets fail to aggregate properly? d. Plasmin proteolysis of cross-linked fibrin
a. Factor VIII
b. Fibrinogen
c. Thrombin
d. VWF
CHAPTER 40 Normal Hemostasis and Coagulation 645

6. What serine protease forms a complex with factor VIIIa, 9. Coagulation factor VIII circulates bound to:
and what is the substrate of this complex? a. VWF
a. Factor VIIa, factor X b. Factor IX
b. Factor Va, prothrombin c. Platelets
c. Factor Xa, prothrombin d. Factor V
d. Factor IXa, factor X
10. Most coagulation factors are synthesized in:
7. What protein secreted by endothelial cells activates a. The liver
fibrinolysis? b. Monocytes
a. Plasminogen c. Endothelial cells
b. TPA d. Megakaryocytes
c. PAI-1
d. TAFI

8. What two regulatory proteins form a complex that digests


activated factors V and VIII?
a. TFPI and serpin
b. Antithrombin and protein C
c. APC and protein S
d. 2-Macroglobulin and 1-antitrypsin

REFERENCES
1. Aird WC: Hemostasis and irreducible complexity. J Thromb Disorders of hemostasis and thrombosis, ed 2, New York, 2001,
Haemost 1:227-230, 2003. McGraw-Hill, pp 3-19.
2. Collen D, Lijnen HR: Tissue-type plasminogen activator: a 14. Dellas C, Loskutoff DJ: Historical analysis of PAI-1 from its
historical perspective and personal account. J Thromb Haemost discovery to its potential role in cell motility and disease.
2:541-546, 2004. Thromb Haemost 93:631-640, 2005.
3. Furie B, Furie BC: Molecular and cellular biology of blood 15. Polgar J, Matuskova J, Wagner DD: The P-selectin, tissue
coagulation. N Engl J Med 326:800-806, 1992. factor, coagulation triad. J Thromb Haemost 3:1590-1596,
4. Ruggeri ZM: Von Willebrand factor, platelets and endothelial 2005.
cell interactions. J Thromb Haemost 1:1335-1342, 2003. 16. Cramer EM, Vainchenker W: Platelet production: cellular and
5. Ruggeri ZM, Savage B: Plateletvessel wall interactions in molecular regulation. In Colman RW, Marder VJ, Clowes AM,
flowing blood. In Colman RW, Marder VJ, Clowes AM, et al, et al, editors: Hemostasis and thrombosis: basic principles and
editors: Hemostasis and thrombosis: basic principles and clinical clinical practice, ed 5, Philadelphia, 2006, Lippincott Williams
practice, ed 5, Philadelphia, 2006, Lippincott Williams & & Wilkins, pp 443-462.
Wilkins, pp 723-736. 17. Fritsma GA: Platelet production and structure. In Corriveau
6. Moncada SP, Palmer RMJ, Higgs AJ: Nitric oxide: physiology, DM, Fritsma GA, editors: Hemostasis and thrombosis in
pathophysiology and pharmacology. Pharmacol Rev 43:109- the clinical laboratory, Philadelphia, 1988, JB Lippincott,
142, 1991. pp 206-228.
7. Huntington JA: Mechanisms of glycosaminoglycan activation 18. George JN, Colman RW: Overview of platelet structure and
of the serpins in hemostasis. J Thromb Haemost 1:1535-1549, function. In Colman RW, Marder VJ, Clowes AM, et al,
2003. editors: Hemostasis and thrombosis: basic principles and clinical
8. Francis CW, Berkowitz SD: Antithrombotic and thrombolytic practice, ed 5, Philadelphia, 2006, Lippincott Williams &
agents. In Kitchens CS, Alving BM, Kessler CM, editors: Wilkins, pp 437-442.
Consultative hemostasis and thrombosis, Philadelphia, 2002, 19. Colman RW, Clowes AW, George JN, et al: Overview of
Saunders, pp 375-393. hemostasis. In Colman RW, Marder VJ, Clowes AM, et al,
9. Haberichter SL, Montgomery RS: Structure and function of editors: Hemostasis and thrombosis: basic principles and clinical
von Willebrand factor. In Colman RW, Marder VJ, Clowes practice, ed 5, Philadelphia, 2006, Lippincott Williams &
AM, et al, editors: Hemostasis and thrombosis: basic principles Wilkins, pp 1-16.
and clinical practice, ed 5, Philadelphia, 2006, Lippincott 20. Walsh PN: Role of platelets in blood coagulation. In Colman
Williams & Wilkins, pp 707-722. RW, Marder VJ, Clowes AM, et al, editors: Hemostasis
10. Bevilacqua MP, Nelson RM: Selectins. J Clin Invest 91:379- and thrombosis: basic principles and clinical practice, ed 5,
387, 1993. Philadelphia, 2006, Lippincott Williams & Wilkins, pp 605-
11. Kansas GS: Selectins and their ligands: current concepts and 616.
controversies. Blood 88:3259-3287, 1996. 21. Mackman N, Tilley RE, Key NS: Role of the extrinsic pathway
12. Morrissey JH, Mutch NJ: Tissue factor structure and function. of blood coagulation in hemostasis and thrombosis. Arterio-
In Colman RW, Marder VJ, Clowes AM, et al, editors: scler Thromb Vasc Biol 27:1687-1693, 2007.
Hemostasis and thrombosis: basic principles and clinical practice, 22. Carlos TM, Harlan JM: Leukocyte-endothelial adhesion mol-
ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins, ecules. Blood 84:2068-2101, 1994.
pp 91-106. 23. Sherry S: The founding of the International Society on
13. Hathaway WE, Goodnight SH: Mechanisms of hemostasis Thrombosis and Haemostasis: how it came about. Thromb
and thrombosis. In Hathaway WE, Goodnight SH, editors: Haemost 64:188-191, 1990.
646 PART VIII Hemostasis and Thrombosis

24. Saito H: Normal hemostatic mechanisms. In Ratnoff OD, 42. Lwaleed BA, Bass PS: Tissue factor pathway inhibitor: struc-
Forbes CD, editors: Disorders of hemostasis, ed 3, Philadelphia, ture, biology, and involvement in disease. J Pathol 208:327-
1996, WB Saunders, pp 23-52. 339, 2006.
25. Coughlin SR: Protease-activated receptors in hemostasis, 43. Monroe DM, Key NS: The tissue factorfactor VIIa complex:
thrombosis and vascular biology. J Thromb Haemost 3:1800- procoagulant activity, regulation, and multitasking. J Thromb
1814, 2005. Haemost 5:1097-1105, 2007.
26. Lane DA, Philippou H, Huntington JA: Directing thrombin. 44. Hakeng TM, Sere KM, Tans G, et al: Protein S stimulates
Blood 106:2605-2612, 2005. inhibition of the tissue factor pathway by tissue factor
27. Mosesson MW: Fibrinogen and fibrin structure and functions. pathway inhibitor. Proc Natl Acad Sci U S A 103:3106-3111,
J Thromb Haemost 3:1894-1904, 2005. 2006.
28. Lorand L: Factor XIII and the clotting of fibrinogen: from 45. Golino P: The inhibitors of the tissue factor:factor VII
basic research to medicine. J Thromb Haemost 3:1337-1348, pathway. Thromb Res 106:V257-V265, 2002.
2005. 46. Dahlback B: The protein C anticoagulant system: inherited
29. Blann AD: Plasma von Willebrand factor, thrombosis, and defects as basis for venous thrombosis. Thromb Res 77:1-43,
the endothelium: the first 30 years. Thromb Haemost 95:49-55, 1995.
2006. 47. de Moerloose P, Bounameaux HR, Mannucci PM: Screening
30. Sadler JE: Von Willebrand factor. J Biol Chem 266:22777- test for thrombophilic patients: which tests, for which patient,
22780, 1991. by whom, when, and why? Semin Thromb Hemost 24:321-327,
31. Kunicki TJ: Platelet glycoprotein polymorphisms and rela- 1998.
tionship to function, immunogenicity, and disease. In 48. Rau JC, Beaulieu LM, Huntington JA, et al: Serpins in throm-
Colman RW, Marder VJ, Clowes AM, et al, editors: Hemostasis bosis, hemostasis and fibrinolysis. J Thromb Haemost 5(suppl
and thrombosis: basic principles and clinical practice, ed 5, 1):102-115, 2007.
Philadelphia, 2006, Lippincott Williams & Wilkins, pp 493- 49. Corral J, Gonzalez-Conejero R, Hernandez-Espinosa D, et al:
506. Protein Z/Z-dependent protease inhibitor (PZ/ZPI) antico-
32. Jacquemin M, De Maeyer M, DOiron R, et al: Molecular agulant system and thrombosis. Br J Haematol 137(2):99-
mechanisms of mild and moderate hemophilia A. J Thromb 108, 2007.
Haemost 1:456-463, 2003. 50. Broze GJ, Jr: Protein Z-dependent regulation of coagulation.
33. Harrison P, Wilborn B, Devilli N, et al: Uptake of plasma Thromb Haemost 86:8-13, 2001.
fibrinogen into the granules of human megakaryocytes and 51. Broze GJ: Protein Z and protein Z-dependent protease inhibi-
platelets. J Clin Invest 84:1320-1324, 1989. tor. In Colman RW, Marder VJ, Clowes AM, et al, editors:
34. Mosesson MW: Fibrinogen and fibrin structure and functions. Hemostasis and thrombosis: basic principles and clinical practice,
J Thromb Haemost 3:1894-1904, 2005. ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins,
35. Broze GJ: Tissue factor pathway inhibitor and the revised pp 215-220.
theory of coagulation. Ann Rev Med 46:103-112, 1995. 52. Kolev K, Machovich R: Molecular and cellular modulation of
36. Roberts HR, Escobar MA: Less common congenital disorders fibrinolysis. Thromb Haemost 89:610-621, 2003.
of hemostasis. In Kitchens CS, Alving BM, Kessler CM, editors: 53. Booth NA, Bachmann F: Plasminogen-plasmin system. In
Consultative hemostasis and thrombosis, ed 2, Philadelphia, Colman RW, Marder VJ, Clowes AM, et al, editors: Hemostasis
2007, Saunders, p 70. and thrombosis: basic principles and clinical practice, ed 5,
37. Corriveau DM: Major elements of hemostasis. In Corriveau Philadelphia, 2006, Lippincott Williams & Wilkins, pp 335-
DM, Fritsma GA, editors: Hemostasis and thrombosis in the 364.
clinical laboratory, Philadelphia, 1988, JB Lippincott, pp 1-33. 54. Bennett B, Ogston D: Fibrinolytic bleeding syndromes. In
38. Monroe DM, Hoffman M: What does it take to make the Ratnoff OD, Forbes CD, editors: Disorders of hemostasis, ed 3,
perfect clot? Arterioscler Thromb Vasc Biol 26:41-48, 2006. Philadelphia, 1996, WB Saunders, pp 296-322.
39. Hoffman M: Remodeling the blood coagulation cascade. 55. Bouma BN, Mosnier LO: Thrombin activatable fibrinolysis
J Thromb Thrombolysis 16:17-20, 2003. inhibitor (TAFI) at the interface between coagulation and
40. Hoffman M, Monroe DM: Coagulation 2006: a modern view fibrinolysis. Pathophysiol Haemost Thromb 33:375-381, 2003/
of hemostasis. Hematol Oncol Clin North Am 21:1-11, 2007. 2004.
41. Hakeng TM, Maurissen LFA, Castoldi E, et al: Regulation of 56. Lowe GD: Circulating inflammatory markers and risks of
TFPI function by protein S. J Thromb Haemost 7(suppl 1):165- cardiovascular and non-cardiovascular disease. J Thromb
168, 2009. Haemost 3:1618-1627, 2005.
Hemorrhagic Coagulation
Disorders
41
George A. Fritsma*

OUTLINE OBJECTIVES
Symptoms of After completion of this chapter, the reader will be able to:
Coagulopathies
Localized versus 1. Distinguish among the causes of localized versus gen- 4. Use the results of laboratory tests to identify and
Generalized Hemorrhage eralized, soft tissue versus mucocutaneous, and monitor the treatment of congenital single coagulation
Mucocutaneous versus acquired versus congenital bleeding. factor deficiencies such as deficiencies of factors VIII,
Anatomic Hemorrhage 2. List laboratory tests to differentiate among acquired IX, and XI.
Acquired versus hemorrhagic disorders of trauma, liver disease, 5. Explain the principle and rationale for the use of each
Congenital Bleeding vitamin K deficiency, and kidney failure, and interpret laboratory test for the detection and monitoring of
Disorders results when given. hemorrhagic disorders.
Acquired Coagulopathies 3. Interpret laboratory assay results to diagnose and 6. Discuss the laboratory monitoring of therapy for hem-
Acute Coagulopathy of subtype von Willebrand disease. orrhagic disorders.
Trauma-Shock
Liver Disease
Renal Failure and
Hemorrhage
Vitamin K Deficiency CASE STUDY
Autoanti-VIII Inhibitor and After studying this chapter, the reader should be able to respond to the following case study:
Acquired Hemophilia
Acquired von Willebrand A 55-year-old man comes to the emergency department with epistaxis (uncontrolled nosebleed).
Disease He reports that he has bleeders disease and has had multiple episodes of bleeding into his joints,
Disseminated Intravascular causing inflammation and hemarthroses. Physical examination reveals swollen, immobilized knees,
Coagulation mild jaundice, and an enlarged liver and spleen. The CBC indicates that the patient is anemic
Congenital and has thrombocytopenia with a platelet count of 74,400/mcL (reference interval, 150,000 to
Coagulopathies 450,000/mcL). The PT is 18 seconds (reference interval, 12 to 14 seconds), and the PTT is
Von Willebrand Disease 43 seconds (reference range, 25 to 35 seconds).
Hemophilia A
1. What is the most likely diagnosis or diagnoses?
Hemophilia B (Factor IX
2. What treatment does the patient need?
Deficiency)
Hemophilia C (Rosenthal
Syndrome, Factor XI
Deficiency)
Other Congenital Single-
Factor Deficiencies

complete patient and family history and do a thorough physi-


SYMPTOMS OF COAGULOPATHIES
cal examination before ordering diagnostic laboratory tests.1
Hemorrhage is a severe form of bleeding that requires interven-
tion. Hemorrhage may be localized or generalized, acquired or Localized versus Generalized Hemorrhage
congenital. To establish the cause of either a bleeding event or Hemorrhage from a single location commonly indicates injury,
a patients tendency to bleed, the physician must obtain a infection, tumor, or an isolated blood vessel defect and is

*The author acknowledges the substantial contributions of Marisa B. Marques, MD, to chapter 41 of the third edition of this textbook, many of
which continue in this edition.

647
648 PART VIII Hemostasis and Thrombosis

called localized bleeding or localized hemorrhage. A surgical site


that bleeds excessively because of inadequate cauterization or BOX 41-1 Generalized Bleeding Signs Heralding a
an ineffective suture is an example of localized bleeding. Local- Possible Hemostatic Defect
ized bleeding seldom implies a defect of vessels, a qualitative Purpurarecurrent, chronic bruising in multiple locations; called
platelet defect, a reduced platelet count, or the coagulation petechiae when less than 3mm in diameter, ecchymoses when
cascade mechanism. greater than 3mm in diameter
Bleeding from multiple sites, spontaneous and recurring Epistaxisnosebleeds that are recurrent, last longer than 10
bleeds, or a hemorrhage that requires physical intervention minutes, or require physical intervention
and transfusions is called generalized bleeding. Generalized Recurrent or excessive bleeding from trauma, surgery, or dental
bleeding is potential evidence for a disorder of primary hemo- extraction
stasis involving defects in blood vessels, defects in platelets, or Bleeding into multiple body cavities, joints, or soft tissue
reduced platelet count, or secondary hemostasis involving defects Simultaneous hemorrhage from several sites
in the coagulation cascade mechanism. Disorders of primary Menorrhagia (menstrual hemorrhage)
or secondary hemostasis that exhibit generalized bleeding or Bleeding that is delayed or recurrent
hemorrhage are called coagulopathies. Bleeding that is inappropriately brisk
Bleeding for no apparent reason
Mucocutaneous versus Hematemesis (vomiting of blood)
Anatomic Hemorrhage
Generalized bleeding may exhibit a mucocutaneous or a soft
tissue (anatomic) pattern. Repeated joint bleeds cause painful inflammation and perma-
Mucocutaneous hemorrhage may be manifested as bruises nent cartilage damage that swells and immobilizes the joint.
or purpura, purple lesions of the skin caused by extravasated Bleeds into soft tissues such as muscle or fat may cause nerve
(seeping) red blood cells (RBCs).2 Petechiae and ecchymoses are compression and subsequent temporary or permanent loss
purpura of less than or greater than 3mm, respectively, and of function.3 When the bleeding involves body cavities, it
the presence of more than one such lesion may indicate a causes symptoms related to the organ that is affected. Bleeding
disorder of primary hemostasis. Other symptoms of defects of into the central nervous system, for instance, may cause
primary hemostasis are menorrhagia (profuse menstrual flow), headaches, confusion, seizures, and coma and must be managed
bleeding from the gums, hematemesis (vomiting of blood), as a medical emergency. Bleeds into the kidney may present
and epistaxis (uncontrolled nosebleed). Although nosebleeds as hematuria and may potentially be associated with acute
are common and mostly innocent, especially in children, they kidney failure.
suggest a primary hemostatic defect when they occur repeat- Whenever a soft tissue or mucocutaneous generalized
edly, last longer than 10 minutes, involve both nostrils, or hemostatic disorder is suspected, hemostasis laboratory testing
require physical intervention or transfusions. is essential. Emergency department physicians often must
Mucocutaneous hemorrhage tends to be associated with begin to intervene in cases of acute hemorrhage, even before
thrombocytopenia (platelet count lower than 150,000/mcL), taking a history or waiting for the results of laboratory assays.
qualitative platelet disorders, von Willebrand disease (VWD), In these instances, group-matched or O-negative RBCs, frozen
or vascular disorders such as scurvy or telangiectasia (see Chap- plasma (FP), platelet concentrate, cryoprecipitate, recombinant
ters 43 and 44). A careful history and physical examination activated coagulation factor VII (rFVIIa; NovoSeven, Novo
distinguishes between mucocutaneous and anatomic bleeding, Nordisk, Inc., Princeton, N.J.) or the antifibrinolytic tranexamic
and the distinction helps direct investigative laboratory testing acid (Cyklokapron, Pfizer, New York, N.Y.) may be used to
and treatment. correct coagulation factor and platelet deficiencies to stop
Anatomic (soft tissue) hemorrhage is seen in acquired or hemorrhage.4-6 Box 41-1 lists symptoms that suggest general-
congenital defects in secondary hemostasis, or plasma coagula- ized hemorrhagic disorders.
tion factor deficiencies. Examples of anatomic bleeding include
recurrent or excessive bleeding after minor trauma, dental Acquired versus Congenital
extraction, or a surgical procedure. In such cases, hemorrhage Bleeding Disorders
may immediately follow a traumatic event, but is often delayed Liver disease, kidney failure, chronic infections, autoimmune
or recurs minutes or hours after the initial blood flow is stopped. disorders, obstetric complications, dietary deficiencies such as
In other patients, anatomic bleeding episodes are spontaneous. vitamin C or vitamin K deficiency, blunt or penetrating trauma,
Most anatomic bleeds are internal, such as bleeds into joints, and inflammatory disorders may all be associated with coagu-
body cavities, muscles, or the central nervous system, and may lopathy. If a patients bleeding episodes began after infancy,
initially have few visible signs. Because joint bleeds cause are associated with some disease or physical trauma, and are
swelling and acute pain, they may not be immediately recog- not duplicated in relatives, they probably are caused by an
nized as hemorrhages, although hemophilia patients usually acquired, not a congenital, condition. When an adult patient
know what is happening from experience. Repeated joint comes for treatment of generalized hemorrhage, the physician
bleeds (hemarthroses) cause painful inflammation, with swell- first looks for an underlying disease or event and takes a per-
ing and permanent cartilage damage that immobilize the joint. sonal and family history. The important elements of patient
CHAPTER 41 Hemorrhagic Coagulation Disorders 649

TABLE 41-1 Screening Tests for a Generalized congenital or inherited coagulopathies. Chronic diseases or
Hemostatic Disorder disorders commonly associated with bleeding are liver disease,
vitamin K deficiency, and renal failure. In all cases, laboratory
Test Assesses for
test results are necessary to confirm the diagnosis and guide
Hemoglobin, hematocrit, Anemia associated with chronic bleeding; the management of acquired hemorrhagic disorders.8
reticulocyte count bone marrow response
Platelet count Thrombocytopenia Acute Coagulopathy of Trauma-Shock
PT Deficiencies of factors II (prothrombin), V, VII, In North America, unintentional injury is the leading cause of
or X (clotting time prolonged) death among those aged 1 to 45 years. The total rises when
PTT Deficiencies of all factors except VII and XIII felonious, self-inflicted, and combat injuries are included: in
(clotting time prolonged) the United States alone, trauma causes 93,000 deaths per year.9
Thrombin time Hypofibrinogenemia and dysfibrinogenemia Severe neurologic displacement accounts for 50% of deaths,
with most occurring before the patient arrives at the hospital;
PT, Prothrombin time; PTT, partial thromboplastin time. however, of initial survivors, 20,000 die of hemorrhage within
48 hours.10 Acute coagulopathy of trauma-shock (ACOTS) accounts
BOX 41-2 Indications of Congenital Hemorrhagic for fatal hemorrhage; 3000 to 4000 of these deaths are prevent-
Disorders able. ACOTS is triggered by the combination of injury-
Relatives with similar bleeding symptoms related acute inflammation, platelet activation, tissue factor release,
Onset of bleeding in infancy or childhood hypothermia, acidosis, and hypoperfusion (poor distribution of
Bleeding from umbilical cord or circumcision wound blood to tissues caused by low blood pressure), all of which
Repeated hemorrhages in childhood, adulthood are elements of systemic shock. For in-transit fluid resuscita-
Hemorrhage into joints, central nervous system, soft tissues, tion, colloid plasma expanders such as 5% dextrose are used
peritoneum to counter hypoperfusion.11 The administration of plasma
expanders has a dilutional effect that is compounded by emer-
gency department transfusion of RBCs, and although these
history are age, sex, current or past pregnancy, any systemic measures are life saving, they intensify the coagulopathy, as
disorders such as diabetes or cancer, trauma, and exposure to does subsequent surgical intervention.
drugs, including over the counter drugs or drugs of abuse. The Massive transfusion, defined as administration of 4 to 5
physician determines the trigger, location, and volume of RBC units within 1 hour or 8 to 10 units within 24 hours, is
bleeding, then orders laboratory assays (Table 41-1). The initial essential in otherwise healthy trauma victims when the systolic
coagulopathy test profile should consist of a complete blood blood pressure is less than 110mmHg, pulse is greater than
count (CBC) that includes a platelet count, prothrombin time 105 beats/min, pH is less than 7.25, hematocrit is less than
(PT), partial thromboplastin time (PTT), and fibrinogen assay.7 32%, hemoglobin is less than 10g/dL, and PT is prolonged to
These tests take on clinical significance when the history and greater than 1.5 times the mean of the reference interval or
physical examination already have established the existence of generates an international normalized ratio (INR) of 1.5.12
abnormal bleeding. They are not effective as screens for healthy
individuals. Management of Acute Coagulopathy
Congenital hemorrhagic disorders are uncommon, occur- of Trauma-Shock
ring in 1 per 100 people, and are usually diagnosed in infants According to the American Society of Anesthesiologists surgical
or young children, who often have relatives with similar symp- practice guidelines, RBC transfusions are required when the
toms. Congenital bleeding disorders lead to repeated hemor- hemoglobin is less than 6.0g/dL and are contraindicated when
rhages that may be spontaneous or occur following minor the hemoglobin is greater than 10.0g/dL.13 For hemoglobin
injury or may occur in unexpected locations, such as joints, concentrations between 6.0 and 10.0g/dL, the decision to
body cavities, retinal veins and arteries, or the central nervous transfuse is based upon the acuity of the patients condition as
system. Patients with mild congenital hemorrhagic disorders determined by physical evidence of blood loss; current blood loss
may have no symptoms until they reach adulthood or experi- rate; blood pressure; arterial blood gas values, especially pH and
ence some physical challenge, such as trauma, dental extrac- oxygen saturation; urine output; and laboratory evidence for coagu-
tion, or a surgical procedure. The most common congenital lopathy, provided there is time for specimens to be collected
deficiencies are VWD, factor VIII and IX deficiencies (hemo- and laboratory assays completed.
philia A and B), and platelet function disorders. Inherited defi- During surgery, coagulopathy is assessed based on microvas-
ciencies of fibrinogen, prothrombin, and factors V, VII, X, XI, cular bleeding, which is evaluated by measuring the volume of
and XIII also exist, but are rare (Box 41-2). blood filling suction canisters, surgical sponges, and surgical
drains. Platelet concentrate is ordered when the platelet count is
less than 50,000/mcL unless the surgeon anticipates limited
ACQUIRED COAGULOPATHIES
blood loss. Platelet concentrate therapy is generally ineffective
Far more patients have acquired bleeding disorders secondary when the patient is known to have immune thrombocytopenic
to trauma, drug exposure, or chronic disease than have purpura, thrombotic thrombocytopenic purpura, or heparin-induced
650 PART VIII Hemostasis and Thrombosis

thrombocytopenia. In patients with these conditions, therapeutic Potential adverse effects of FP administration are transfusion-
platelets are rapidly consumed, and their administration may related acute lung injury (TRALI) and TACO. The effects of TRALI
therefore be contraindicated, although they may provide tem- and TACO both can lead to potentially fatal acute adult respira-
porary rescue. Platelet concentrate is never ordered when the tory distress syndrome. FP therapy may also cause anaphylaxis,
platelet count is greater than 100,000/mcL. Platelet administra- thrombosis, and multiple organ failure.
tion may be necessary when the platelet count is between
50,000 and 100,000/mcL provided there is bleeding into a Use of Activated Prothrombin Complex Concentrate,
confined space such as the brain or eye; if the patient is taking Recombinant Activated Factor VII, and Tranexamic
aspirin or clopidogrel; if there is a known platelet disorder such Acid in Acute Coagulopathy of Trauma-Shock
as a release defect, Glanzmann thrombasthenia, or Bernard- When administration of RBCs, platelets, cryoprecipitate, and
Soulier syndrome (see Chapter 43); or if the surgery involves FP fails to achieve hemostasis, activated prothrombin complex
cardiopulmonary bypass, which suppresses platelet activity. concentrate (FEIBA [factor eight inhibitor bypassing activity],
Baxter Healthcare Corp., Deerfield, Ill.; Autoplex T, Nabi Bio-
Use of Frozen Plasma in Acute Coagulopathy pharmaceuticals, Inc., Boca Raton, Fla.), recombinant activated
of Trauma-Shock coagulation factor VII (NovoSeven), or the antifibrinolytic drug
FP is thawed, warmed to 37 C, and transfused when there is tranexamic acid (Cyklokapron) may be employed. Autoplex T
microvascular bleeding and the PT is prolonged to greater than or FEIBA may be used at a dosage of 50 units/kg every 12 hours
1.5 times the mean of the PT reference interval or if the PTT is not to exceed 200 units/kg in 24 hours. The dose-response
prolonged to greater than 2 times the mean of the PTT refer- relationship of FEIBA is variable, and because FEIBA contains
ence interval. FP is also transfused when the patient is known activated coagulation factors it raises the risk of disseminated
to have a preexisting single coagulation factor deficiency such intravascular coagulation (DIC). NovoSeven has been cleared
as factor VIII deficiency and no factor concentrate is available, by the Food and Drug Administration (FDA) for use in treating
when there has been significant volume replacement with hemophilia A or B when factor VIII or factor IX inhibitors are
colloid plasma expanders and RBCs, and when hemorrhage is present, respectively; its application in the treatment of ACOTS
caused by warfarin overdose (see Chapter 46). is off label. A NovoSeven dosage of 30mcg/kg is instantly
FP is used only to treat coagulopathy and should not effective in stopping microvascular hemorrhage in nonhemo-
be used as a blood volume expander. The dosage is 10 to philic trauma victims, and NovoSeven does not cause DIC.17,18
15mL/kg in continuous infusion. Theoretically, FP should be However, one study found a possible link between off-label
transfused until 30% coagulation factor activity has been NovoSeven use and arterial and venous thrombosis in patients
reached for all factors; however, the actual volume adminis- with existing thrombotic risk factors.19 Cyklokapron may be
tered is limited by the risk of transfusion-associated circulatory used at a loading dose of 1g administered over 10 minutes
overload (TACO). followed by infusion of 1g over 8 hours. First approved in
Although many refer to frozen plasma as fresh frozen 1986 to prevent bleeding in hemophilic patients who are about
plasma, this term no longer applies, because most FP is pre- to undergo invasive procedures, Cyklokapron is effective for
pared up to 24 hours after the time of collection rather than ACOTS, but this is an off-label use.
within 8 hours, as was the traditional practice.14 High-volume No laboratory tests are available to monitor the concentra-
trauma centers such as combat facilities may maintain a small tions of Autoplex T, FEIBA, NovoSeven, or Cyklokapron. Labo-
inventory of thawed FP ready for administration. Thawed FP ratory directors characteristically advise surgeons to monitor
may be stored at 1 C to 6 C for up to 5 days subsequent to the effectiveness of all ACOTS therapy indirectly by checking
thawing. The activity of von Willebrand factor (VWF) and for the correction of platelet count, PT, and PTT to within their
coagulation factors V and VIII decline to approximately 60% respective reference intervals. Platelet aggregometry may be
after 5 days of refrigerator storage; however, the product may used to establish platelet function efficacy, and coagulation
still be transfused.15 factor assays are valuable as follow-up assays to PT and PTT to
Administration of cryoprecipitate is indicated when there is determine if the target activity of 30% has been met. Once the
microvascular bleeding and the fibrinogen concentration is less condition of patients with ACOTS has been stabilized, addi-
than 100mg/dL. A 15- to 20-mL unit of cryoprecipitate pro- tional hemostasis-related therapy is seldom required.
vides 150 to 250mg of fibrinogen, and the risk of TACO is
lower than that with FP or plasma expanders. Often dosages Liver Disease
of RBCs, platelets, FP, and cryoprecipitate are estimated in the The bleeding associated with liver disease may be localized
absence of laboratory monitoring when the patients condition or generalized, mucocutaneous or anatomic. Enlarged and
is emergent. collateral esophageal vessels called esophageal varices are a
Most trauma center transfusion service directors require sur- complication of chronic alcoholic cirrhosis; hemorrhaging
geons and emergency department physicians to transfuse 1 from varices is localized bleeding, not a coagulopathy, though
unit of FP for every 4 units of RBCs, although there is battlefield often fatal. Mucocutaneous bleeding occurs in liver disease
evidence that a ratio of 1 unit of FP per single unit of RBCs associated thrombocytopenia, often accompanied by decreased
may lead to more favorable mortality statistics.16 The latter platelet function. Soft tissue bleeding is the consequence of
approach places a strain on the FP supply, however. procoagulant dysfunction and deficiency.
CHAPTER 41 Hemorrhagic Coagulation Disorders 651
Procoagulant Deficiency in Liver Disease production of regulatory antithrombin, protein C, or protein
The liver produces nearly all of the plasma coagulation factors S and by the release of activated procoagulants from degenerat-
and regulatory proteins. Hepatitis, cirrhosis, obstructive jaun- ing liver cells. In addition, the failing liver cannot clear acti-
dice, cancer, poisoning, and congenital disorders of bilirubin vated coagulation factors that are regularly produced by
metabolism may suppress the synthetic function of hepato- abdominal organs and transported through the portal circula-
cytes, reducing either the concentrations or activities of the tion. In primary or metastatic liver cancer, hepatocytes also
plasma coagulation factors to below hemostatic levels (less may produce procoagulant substances that trigger chronic DIC,
than 30% of normal). leading to ischemic complications.
Liver disease predominantly alters the production of the In acute, uncompensated DIC, the PT, PTT, and thrombin
vitamin Kdependent factors II (prothrombin), VII, IX, and X time are prolonged; the fibrinogen level is reduced to less
and control proteins C, S, and Z. In liver disease, these seven than 100mg/dL; and fibrin degradation products, including
factors are produced in their des--carboxyl forms, which cannot D-dimers, are significantly increased.27 If the DIC is chronic and
participate in coagulation (see Chapter 40). At the onset of liver compensated, the only elevated test result may be the D-dimer
disease, factor VII, which has the shortest plasma half-life at 6 assay value, a hallmark of unregulated coagulation and fibrino-
hours, is the first coagulation factor to exhibit decreased activ- lysis.28 Although DIC can be resolved only by removing its
ity. Because the PT is particularly sensitive to factor VII activity, underlying cause, its hemostatic deficiencies may be temporar-
it is characteristically prolonged in mild liver disease, serving ily corrected by administering RBCs, FP, platelet concentrates,
as a sensitive early marker.20 Vitamin K deficiency caused by activated protein C, or antithrombin concentrates.29,30
dietary insufficiency independent of liver disease produces a
similar effect on the PT (see the case study in this chapter). Hemostasis Laboratory Tests in Liver Disease
Declining coagulation factor V activity is a more specific The PT, PTT, thrombin time, fibrinogen concentration, platelet
marker of liver disease than deficient factor II, VII, IX or X count, and D-dimer concentration are useful in characterizing
because factor V is nonvitamin K dependent and is not affected the hemostatic abnormalities in liver disease (Table 41-2).
by dietary vitamin K deficiency. The factor V activity assay,
performed in conjunction with the factor VII assay, may be
used to distinguish liver disease from vitamin K deficiency.21 TABLE 41-2 Hemostasis Laboratory Tests in Liver
Fibrinogen is an acute phase reactant that is frequently Disease
elevated in early or mild liver disease. Moderately and severely Assay Interpretation
diseased liver produces fibrinogen that is coated with excessive
Fibrinogen >400mg/dL in early, mild liver disease;
sialic acid, a condition called dysfibrinogenemia in which the
<200mg/dL in moderate to severe liver
fibrinogen functions poorly. Dysfibrinogenemia causes gener-
disease causing hypofibrinogenemia or
alized soft tissue bleeding associated with a prolonged throm-
dysfibrinogenemia
bin time and an exceptionally prolonged reptilase clotting
Thrombin time Prolonged in dysfibrinogenemia,
time.22 In end-stage liver disease, the fibrinogen level may fall
hypofibrinogenemia, elevation of fibrin
to less than 100mg/dL, which is a mark of liver failure.23
degradation products, and therapy with
VWF and factors VIII and XIII are acute phase reactants that
unfractionated heparin
may be unaffected or elevated in mild to moderate liver
Reptilase time Prolonged in hypofibrinogenemia,
disease.24,25 In contrast to the other coagulation factors, VWF is
significantly prolonged in
produced from endothelial cells and megakaryocytes and is
dysfibrinogenemia; assay rarely used
stored in endothelial cells and platelets.
PT Prolonged even in mild liver disease due
to presence of des--carboxyl factors in
Platelet Abnormalities in Liver Disease
place of normal factors II (prothrombin),
Moderate thrombocytopenia occurs in a third of patients with
VII, IX, and X. Report PT in seconds,
liver disease. Platelet counts of less than 150,000/mcL may
not INR, when testing for liver disease.
result from sequestration and shortened platelet survival asso-
PTT Mildly prolonged in severe liver disease
ciated with portal hypertension and resultant hepatospleno-
due to DIC or des--carboxyl factors II
megaly. In alcoholism-related hepatic cirrhosis, acute alcohol
(prothrombin), IX, and X
toxicity also suppresses platelet production. Platelet aggrega-
Platelet count Mild thrombocytopenia, platelet count
tion and secretion properties are often suppressed; this is
<150,000/mcL
reflected in reduced platelet aggregometry and lumiaggregome-
Platelet aggregometry Mild suppression of platelet aggregation
try results. Occasionally platelets are hyperreactive. Aggregome-
and secretion to most agonists
try may be used to predict bleeding and thrombosis risk.26
D-dimer >240ng/mL by quantitative assay
Euglobulin clot lysis time Lysis in <2hr in primary or secondary
Disseminated Intravascular Coagulation
systemic fibrinolysis; assay rarely used
in Liver Disease
Chronic or compensated DIC (see Chapter 42) is a significant DIC, Disseminated intravascular coagulation; INR, international normalized ratio; PT,
complication of liver disease that is caused by decreased liver prothrombin time; PTT, partial thromboplastin time.
652 PART VIII Hemostasis and Thrombosis

Factor V and VII assays may be used in combination to dif- to moderate mucocutaneous bleeding.32 Platelet adhesion to
ferentiate liver disease from vitamin K deficiency. Both factors blood vessels and platelet aggregation are suppressed, perhaps
are decreased in liver disease, but only factor VII is decreased because the platelets are coated by guanidinosuccinic acid or
in vitamin K deficiency. dialyzable phenolic compounds.33 Decreased RBC mass (anemia)
Plasminogen deficiency, a shortened euglobulin lysis time, and thrombocytopenia contribute to the bleeding and may be
and an elevated D-dimer confirm systemic fibrinolysis. The corrected with dialysis, erythropoietin or RBC transfusions, and
reptilase time occasionally may be useful to confirm dysfibrino- interleukin-11 therapy.34,35
genemia. This test duplicates the thrombin time test except that Hemostasis activation syndromes that deposit fibrin in the
venom of the reptile Bothrops atrox (common lancehead viper) renal microvasculature reduce the function of the glomeruli.
is substituted for thrombin reagent. The Bothrops venom trig- Examples of such disorders are DIC, hemolytic uremic syndrome,
gers fibrin polymerization by cleaving fibrinopeptide A, but and thrombotic thrombocytopenic purpura. Although these
not fibrinopeptide B, from the fibrinogen molecule. The sub- are by definition thrombotic disorders, they invariably cause
sequent polymerization is slowed by structural defects, which thrombocytopenia, which may lead to mucocutaneous bleed-
prolong the interval from reagent addition to clot formation. ing. Fibrin also may be deposited in renal transplant rejection
The reptilase time test is unaffected by standard unfractionated and in the glomerulonephritis syndrome of systemic lupus
heparin therapy and can be used to assess fibrinogen function erythematosus. This may be associated with a rise in quantita-
even when there is heparin in the sample. tive plasma D-dimer, thrombin-antithrombin complexes, or
prothrombin fragment 1 + 2, which are markers of coagulation
Hemostatic Treatment to Resolve Liver activation (see Chapter 45).36
DiseaseRelated Hemorrhage Laboratory tests for bleeding in renal disease provide only
Oral or intravenous vitamin K therapy may correct the bleeding modest information with little predictive or management
associated with des--carboxyl factors II (prothrombin), VII, IX, value. The bleeding time may be prolonged, but is too unreli-
and X, although the therapeutic effect of vitamin K is short- able to provide an accurate diagnosis or to assist in monitoring
lived compared to its effect in dietary vitamin K deficiency treatment.37 Platelet aggregometry test results may predict
because of the livers impaired synthetic ability. In severe liver bleeding. The PT and PTT are expected to be normal.
disease, FP transfusion provides all of the coagulation factors Management of renal failurerelated bleeding typically
in hemostatic concentrations, although VWF and factors V and focuses on the severity of the hemorrhage without reliance on
VIII may be reduced. Owing to the small concentration and laboratory test results. Renal dialysis improves platelet func-
short half-life of factor VII, FP is unlikely to return the PT to tion, particularly when anemia is well controlled. Desmopres-
within the reference interval. sin acetate may be administered intravenously (DDAVP) or
A unit of FP consists of 200 to 280mL. The typical adult FP intranasally (Stimate) to increase the plasma concentration of
dose for liver disease is 2 units, but the dose varies widely high-molecular-weight multimers of VWF, which also aids
depending on the indication and the ability of the patients platelet adhesion and aggregation.38 Patients with renal failure
cardiac and renal system to rapidly excrete excess fluid. TACO should not take aspirin, clopidogrel, or other platelet inhibi-
is likely to occur when 30mL/kg has been administered, but tors, because these drugs increase the risk of hemorrhage.
it may occur with even smaller volumes in patients with com-
promised cardiac or kidney function. Nephrotic Syndrome and Hemorrhage
If the fibrinogen level is less than 50mg/dL, spontaneous Nephrotic syndrome is a state of increased glomerular per
bleeding is imminent, and cryoprecipitate is preferred for its meability associated with a variety of conditions, such as
smaller volume and high fibrinogen concentration, 150 to chronic glomerulonephritis, diabetic glomerulosclerosis, sys-
250mg in a 15- to 20-mL unit. FP and cryoprecipitate present temic lupus erythematosus, amyloidosis, and renal vein throm-
a theoretical risk of virus transmission, as do other untreated bosis.39 In nephrotic syndrome, low-molecular-weight proteins
single-donor biologic blood products. Allergic transfusion are lost through the glomerulus into the glomerular filtrate and
reactions are more common with plasma-containing products. the urine. Coagulation factors II (prothrombin), VII, IX, X and
Other therapeutic options for patients with liver disease XII have been detected in the urine, as have the coagulation
related bleeding are platelet concentrates, prothrombin regulatory proteins antithrombin and protein C. In 25% of
complex concentrate, activated protein C (Xigris, Eli Lilly and cases, loss of regulatory proteins takes precedence over loss
Co., Indianapolis, Ind.), antithrombin concentrate (Throm- of procoagulants and leads to a tendency toward venous
bate III, Telacris Biotherapeutics, Inc., Research Triangle Park, thrombosis.40
N.C.), NovoSeven, and Cyklokapron.
Vitamin K Deficiency
Renal Failure and Hemorrhage Vitamin K is ubiquitous in foods, especially green leafy vege-
Acute renal failure may be associated with acute gastrointesti- tables, and the daily requirement is small, so pure dietary
nal bleeding caused by specific anatomic lesions, but this deficiency is rare. Body stores are limited, however, and become
does not necessarily indicate that a coagulopathy or platelet exhausted when the usual diet is interrupted, as when patients
dysfunction is present.31 Chronic renal failure of any cause are fed only with parenteral (intravenous) nutrition for an
is commonly associated with platelet dysfunction and mild extended period or when patients embark upon fad diets. Also,
CHAPTER 41 Hemorrhagic Coagulation Disorders 653
because vitamin K is fat soluble and requires bile salts for COOH HOOC COOH
absorption, biliary duct obstruction (atresia), fat malabsorp-
tion, and chronic diarrhea may cause vitamin K deficiency.
CH2 CH
Broad-spectrum antibiotics that disrupt normal flora may
GLU GLA
cause a slight reduction because they destroy bacteria that
produce vitamin K. The degree of reduction is insignificant CH2 CH2
when the diet is otherwise normal, but may become an
important issue when the patient is receiving only parenteral
H2N-CH-COOH H2N-CH-COOH
nutrition. -carboxylase

OH O
Hemorrhagic Disease of the Newborn Caused
by Vitamin K Deficiency CH3 CH3
O
Because of their sterile intestines and the minimal concentra-
R R
tion of vitamin K in human milk, newborns are constitution-
OH O
ally vitamin K deficient.41 Hemorrhagic disease of the newborn Vitamin K
Hydroquinone antagonists Epoxide
was common in the United States before routine administra-
tion of vitamin K to infants was legislated in the 1960s, and it
still occurs in developing countries. The activity levels of factors NADP Epoxide
O
reductase
II (prothrombin), VII, IX, and X are lower in normal newborns CH3
Quinone
than in adults, and premature infants have even lower concen-
reductase
trations of these factors.42 Breastfeeding prolongs the defi- R
ciency, because passively acquired maternal antibodies delay NADPH O
the establishment of gut flora. Vitamin K
Figure 41-1 Glutamic acid (GLU) gains a carboxyl group to become
Vitamin K Antagonists -carboxyglutamic acid (GLA) when catalyzed by -carboxylase. This post-
The -carboxylation cycle of coagulation factors is interrupted translational modification enables the vitamin Kdependent coagulation
by coumarin-type oral anticoagulants such as warfarin that factors II (prothrombin), VII, IX, and X and proteins C, S, and Z to bind ionic
disrupt the vitamin K epoxide reductase and vitamin K quinone calcium necessary for normal coagulation. Vitamin K donates the carboxyl
reductase reactions (Figure 41-1). In deficient -carboxylation, group through the quinone reductase pathway. Warfarin inactivates quinone
the liver releases dysfunctional des--carboxyl factors II (pro- reductase and epoxide reductase to prevent carboxylation. NADP, Nicotin-
amide adenine dinucleotide phosphate; NADPH, reduced form of nicotin-
thrombin), VII, IX, and X, and proteins C, S, and Z; these inac-
amide adenine dinucleotide phosphate; R, any side chain.
tive forms are called proteins in vitamin K antagonism (PIVKA).
Therapeutic overdose or the accidental or felonious adminis-
tration of warfarin-containing rat poisons may result in moder- bleeding, FP, Autoplex T, FEIBA, or NovoSeven may be admin-
ate to severe hemorrhage because of the lack of functional istered. There is no direct laboratory assay for FP, Autoplex T,
factors. The effect of brodifacoum or superwarfarin, often used FEIBA, or NovoSeven, but the patients recovery may be moni-
as a rodenticide, lasts for weeks to months, and treatment of tored indirectly using the PT, with an attempt made to restore
poisoning with this substance requires repeated administration the INR to the therapeutic range of 2.0 to 3.0.
of vitamin K with follow-up PT monitoring.43 Warfarin over-
dose is the single most common reason for hemorrhage- Autoanti-VIII Inhibitor and
associated emergency department visits. Acquired Hemophilia
Acquired autoantibodies that specifically inhibit factors II (pro-
Detection of Vitamin K Deficiency or Proteins thrombin), V, VIII, IX, and XIII and VWF have been described
in Vitamin K Antagonism in nonhemophilic patients.44 Anti-VIII is the most common.
Clinical suspicion of vitamin K deficiency is supported by a Patients who develop an autoantibody to factor VIII, which is
prolonged PT with or without a prolonged PTT. In PT and PTT diagnostic of acquired hemophilia, are frequently older than
mixing studies, if pooled platelet-free normal plasma (NP; age 60 and have no apparent underlying disease. Acquired
Cryocheck, Precision BioLogic, Inc., Dartmouth, Nova Scotia) hemophilia is occasionally associated with rheumatoid arthri-
is combined with patient plasma, the mixture yields normal tis, inflammatory bowel disease, systemic lupus erythematosus,
(corrected) PT and PTT results, which indicates that factor defi- or lymphoproliferative disease. It also may occur in women
ciencies were the cause of the prolonged screening test times. of childbearing age 2 to 5 months after delivery. Patients
Specific single-factor assays always detect low factor VII because with inhibitor autoantibodies are given immunosuppressive
of its short half-life, followed in turn by decreases in factors IX, therapy, although autoantibodies that develop after pregnancy
X, and II (prothrombin). typically disappear spontaneously. Acquired hemophilia has
The standard therapy for vitamin K deficiency is oralor, an overall incidence of 1 per 1 million people per year. Patients
in an emergency, intravenousvitamin K. Because synthesis of experience sudden and severe bleeding in soft tissues or bleed-
functional factors requires at least 3 hours, in the case of severe ing in the gastrointestinal or genitourinary tract. Acquired
654 PART VIII Hemostasis and Thrombosis

hemophilia, even when treated, remains fatal in at least 20% to factor VIII (see Hemophilia A and Factor VIII Inhibitors).
of cases. Autoantibodies to other procoagulants are less fre- Titer results help the clinician choose the proper therapy to
quent, but create similar symptoms.45 control bleeding. Repeat titers are used to follow the response
to immunosuppressive drugs, but are not needed for manage-
Clot-Based Studies in Acquired Hemophilia ment of the bleeding symptoms.
PT, PTT, and thrombin time are recommended tests for any
patient with sudden onset of anatomic hemorrhage resembling Other Factor Inhibitors
acquired hemophilia. In the presence of a factor VIII inhibitor, Anticoagulation factor II (antiprothrombin) antibodies,
which is the most common type, the PTT should be prolonged, detectable by immunoassay, develop in approximately 30% of
whereas the PT and thrombin time are expected to be normal. patients with lupus anticoagulant.47 Although lupus anticoagu-
A factor assay should reveal factor VIII activity to be less than lant is associated with thrombosis, some patients with antipro-
30% and often undetectable. thrombin antibodies experience bleeding and have a prolonged
The presence of the inhibitor is confirmed with clot-based PT. A finding of reduced activity on prothrombin assay and a
mixing studies. The PTT prolongation may be corrected ini- positive test result for lupus anticoagulant confirm the diagno-
tially by the addition of NP to the test specimen in a 1:1 ratio, sis. Antiprothrombin antibodies not associated with lupus
but PTT again becomes prolonged after incubation of the 1:1 anticoagulant are rare. Factor V and XIII inhibitors have been
NPpatient plasma mixture at 37 C for 1 to 2 hours. The documented in patients receiving isoniazid treatment for
return of the lack of correction after incubation occurs because tuberculosis.48,49 Antibodies to factor IIa (thrombin) and factor
factor VIII autoantibodies are frequently of the immunoglobu- V may arise after exposure to topical bovine thrombin or fibrin
lin G4 isotype, which is time and temperature dependent. glue.50 Autoanti-X antibodies are rare; however, factor X defi-
Consequently, the inhibitor effect may be evident only after ciency in amyloidosis may be caused by what seems to be an
the patients inhibitor is allowed to interact with the factor VIII absorptive mechanism.51 In many cases of acquired inhibitors,
in the NP for 1 to 2 hours at 37 C before testing. A few high- mixing studies show uncorrected prolongation without incu-
avidity inhibitors may cause immediate prolongation of the bation (immediate mixing study), and inhibitor titers may be
PTT; in this case an incubated mixing study is unnecessary. determined by the Bethesda procedure.
The in vitro kinetics of factor VIII neutralization by inhibitor
are nonlinear. Although there is early rapid loss of factor VIII Management of Acquired Hemophilia
activity, residual activity remains, which indicates that the reac- Autoplex T, FEIBA, or NovoSeven may bypass the acquired
tion has reached equilibrium. This is called type II kinetics coagulation factor VIII or factor IX inhibitor in acquired hemo-
(Figure 41-2). In contrast, alloantibodies to factor VIII, which philia and control acute bleeding. Patients who have low titers
develop in 20% to 25% of patients with severe hemophilia in of inhibitor (less than 5 Bethesda units) may respond to
response to factor VIII therapy, exhibit type I kinetics. In the administration of desmopressin acetate (DDAVP) or factor VIII
latter, there is linear in vitro neutralization of factor VIII activity concentrates, but close monitoring of their response to therapy
over 1 to 2 hours, which results in complete inactivation. In with serial coagulation factor VIII activity assays is warranted.
type I kinetics in vitro measurement is relatively accurate, Once bleeding is controlled, immunosuppressive therapy may
whereas in type II kinetics the titration of inhibitor activity is reduce the inhibitor titer.52 Plasma exchange may also be used
semiquantitative.46 in severe cases, but the response is less reliable than the
Quantitation of autoanti-VIII inhibitor is accomplished response to immunosuppression.
using the Bethesda assay, which is ordinarily employed to
measure inhibitor in hemophilic patients with alloantibodies Acquired von Willebrand Disease
Acquired VWF deficiency, with symptoms similar to those of
congenital VWD, has been described in association with hypo-
thyroidism; autoimmune, lymphoproliferative, and myelopro-
Type I kinetics Type II kinetics liferative disorders; benign monoclonal gammopathies; Wilms
alloantibodies autoantibodies
Factor activity

Factor activity

tumor; intestinal angiodysplasia; congenital heart disease; pes-


ticide exposure; and hemolytic uremic syndrome.53 The patho-
genesis of acquired VWD varies and may involve decreased
production of VWF, presence of an autoantibody, or adsorp-
2 Hours 2 Hours tion of VWF to abnormal cell surfaces, as seen in association
with lymphoproliferative disorders and Wilms tumor.54
Figure 41-2 In type I linear kinetics, the inhibitor fully inactivates the
Acquired VWD manifests with moderate to severe mucocu-
factor in vitro. This is typical of alloantibodies such as factor VIII inhibitors in
taneous bleeding and may be suspected in any patient with
patients with severe hemophilia A. In type II kinetics, the inhibitor and factor
reach equilibrium. This is typical of autoantibodies such as the factor VIII recent onset of bleeding who has no significant medical history.
inhibitor in acquired hemophilia A. Because of the type II kinetics, the Although the PT is not affected, the PTT may be moderately
Bethesda titer is considered a semiquantitative measure in acquired hemo- prolonged if the VWF reduction is severe enough to cause a
philia, although the results are commonly used to distinguish low-titer from deficiency of coagulation factor VIII, for which VWF serves as
high-titer antibodies. a carrier molecule. As in congenital VWD, the diagnosis is
CHAPTER 41 Hemorrhagic Coagulation Disorders 655

based on a finding of diminished VWF activity (ristocetin


Binds platelet site Carries coagulation
cofactor [VWF:RCo] assay) and the presence of VWF antigen GP Ib/IX/V factor VIII
(VWF:Ag) by immunoassay. It may be difficult to differentiate
between mild, previously asymptomatic congenital VWD and
acquired VWD. VWF VIII
If the patient requires treatment for bleeding, DDAVP or a
VWF:Ag
plasma-derived factor VIII/VWF concentrate such as Humate-P
epitope
(CSL Behring, King of Prussia, Pa.), Wilate (Octapharma,
Collagen binding
Hoboken, N.J.), or Alphanate (Grifols, Los Angeles, Calif.) is sites
effective at controlling the symptoms. Cryoprecipitate is no
longer recommended for treatment of VWD because it does Figure 41-3 Large von Willebrand factor (VWF) multimers bind intimal
not undergo viral inactivation. collagen upon desquamation of endothelial cells or more extensive injury and
expand under shear force to form a carpet to which platelets adhere
Disseminated Intravascular Coagulation through platelet glycoprotein (GP) Ib/IX/V binding sites. VWF binds and sta-
bilizes factor VIII, serving as a carrier protein. The VWF antigen (VWF:Ag) is
DIC, although characteristically identified through hemor-
exploited by monoclonal quantitative immunoassays.
rhagic symptoms, is classified as a thrombotic disorder and is
described in Chapter 42.
TABLE 41-3 Nomenclature Relating to the VIII/VWF
CONGENITAL COAGULOPATHIES Complex
von Willebrand Disease Term Meaning
VWD is a common mucocutaneous bleeding disorder first
VIII/VWF Customary term for the plasma combination of factor VIII
described by Finnish Professor Erik von Willebrand in 1926.
and VWF.
VWD is caused by any one of dozens of germline mutations
VIII Procoagulant factor VIII, the protein transported on VWF.
that result in quantitative or structural abnormalities of VWF.
Factor VIII binds activated factor IX to form the complex
Both quantitative and structural abnormalities lead to decreased
of VIIIa-IXa, which digests and activates factor X. Factor
adhesion by platelets to injured vessel walls, causing impaired
VIII deficiency is called hemophilia A.
primary hemostasis. VWD is the most prevalent of the congeni-
VWF:Ag Epitope that is the antigenic basis for the VWF
tal bleeding disorders and is found in approximately 1% of the
immunoassay.
population. It affects both sexes because of its autosomal dom-
VWF:RCo Ristocetin cofactor activity, also called VWF activity. VWF
inant inheritance pattern. The parameters of and laboratory
activity is measured by the ability of ristocetin to cause
testing guidelines for VWD are established and defined in the
agglutination of reagent platelets by the patients VWF.
National Heart, Lung, and Blood Institute (NHLBI) publica-
VIII:C Factor VIII coagulant activity as measured in factor-specific
tion The Diagnosis, Evaluation, and Management of von Wille-
clot-based assays.
brand Disease.55

Molecular Biology and Functions of


von Willebrand Factor Each VWF monomer has four functional domains that bind
VWF is a glycoprotein whose molecular mass ranges from factor VIII, platelet glycoprotein Ib/V/IX, platelet glycoprotein
800,000 to 20,000,000D, the largest molecule in human IIb/IIIa, and collagen, respectively (see Chapter 13).59 Upon
plasma. Its plasma concentration is 0.5 to 1.0mg/dL, but a release from intracellular stores, VWF forms a complex with
great deal more is readily available on demand from storage coagulation factor VIII named VIII/VWF (Figure 41-3). VWF
organelles. VWF is synthesized in the endoplasmic reticulum protects factor VIII from proteolysis, prolonging its plasma
of endothelial cells and stored in cytoplasmic Weibel-Palade half-life from a few minutes without VWF to 8 to 12 hours with
bodies of endothelial cells. It is also synthesized in megakaryo- VWF. Table 41-3 lists the nomenclature for the structural and
cytes and stored in the -granules of platelets (see Chapter 13). functional components of the factor VIII/VWF molecule.
Weibel-Palade bodies and -granules release VWF in response Although VWF is the factor VIII carrier molecule, its primary
to a variety of hemostatic stimuli.56 function is to mediate platelet adhesion to subendothelial col-
The VWF gene consists of 52 exons spanning 178 kilobase lagen in areas of high flow rate and high shear force, as in
pairs (kb) on chromosome 12.57 The translated protein is a capillaries and arterioles. VWF first binds fibrillar intimal col-
monomer of 2813 amino acids that, after glycosylation, forms lagen exposed during the desquamation of endothelial cells.
dimers that are transferred to the aforementioned storage Subsequently, platelets adhere through their glycoprotein Ib/V/
organelles, where the dimers polymerize to form enormous IX receptor to the VWF carpet. The largest VWF multimers are
multimers. At the time of storage, a signal sequence and best equipped to serve the adhesion function. When VWF
a propeptide, known as VWF antigen II, are cleaved so that binds glycoprotein Ib/V/IX, platelets become activated and
the mature monomers, already polymerized, consist of 2050 express a second VWF binding site, glycoprotein IIb/IIIa. This
amino acids.58 receptor binds VWF and fibrinogen to mediate irreversible
656 PART VIII Hemostasis and Thrombosis

platelet-to-platelet aggregation. The adhesion and aggregation Application


sequences are essential to normal hemostasis.
Large
Pathophysiology of von Willebrand Disease
Structural (qualitative) or quantitative VWF abnormalities
reduce platelet adhesion, which leads to mucocutaneous hem-
orrhage of varying severity: epistaxis, ecchymosis, menorrha-
gia, gastrointestinal tract bleeding, and surgical bleeding.
Symptoms vary over time and within kindreds, because VWF
production and release is susceptible to a variety of physiologic Small
influences. Severe quantitative VWF deficiency creates in addi-
tion factor VIII deficiency owing to the inability to protect N VWD 1 2A 2B 3
nonbound factor VIII from proteolysis. Many people have VWF Figure 41-4 Schematic of von Willebrand factor multimer analysis by
levels in the intermediate range of 30% to 50% of normal and polyacrylamide gel electrophoresis shows diminished concentration but
maintain a factor VIII level sufficient for competent coagula- normal ratios in von Willebrand disease (VWD) type 1, absence of large and
tion. When factor VIII levels decrease to less than 30% of intermediate multimers in subtype 2A, absence of large forms in subtype 2B,
normal, anatomic soft tissue bleeding accompanies the typical and absence of all multimers in type 3.
mucocutaneous bleeding pattern of VWD.
intermediate-molecular-weight multimers. There also may be
von Willebrand Disease Types and Subtypes moderate thrombocytopenia caused by chronic platelet activa-
Type 1 von Willebrand Disease. Type 1 VWD is a tion, because multimer-coated platelets indiscriminately bind
quantitative VWF deficiency caused by one of several auto the endothelium.
somal dominant frameshifts, nonsense mutations, or dele- A platelet mutation that increases glycoprotein Ib/V/IX
tions.60,61 Type 1 is seen in approximately 75% of VWD patients. affinity for normal VWF multimers creates a clinically similar
The plasma concentrations of VWF and factor VIII are variably, disorder called platelet-type VWD or pseudo-VWD. In this
although proportionally, reduced.62 There is mild to moderate instance, the large multimers also are lost from the plasma, and
bleeding, usually following a hemostatic challenge such as platelets become adhesive (Figure 41-4). Clinically and in the
dental extraction or surgery. In women, menorrhagia is a laboratory, the two entities are indistinguishable.
common complaint that leads to the diagnosis of VWD.
Subtype 2M von Willebrand Disease. Subtype 2M
Type 2 von Willebrand Disease. Type 2 VWD com- VWD is caused by a qualitative variant of VWF that has
prises a variety of qualitative VWF abnormalities. VWF levels decreased platelet receptor binding but a normal multimeric
may be normal or moderately decreased, but VWF function is pattern in electrophoresis. The distinguishing feature of subtype
consistently reduced. 2M that separates it from type 1 is a discrepancy between the
concentration of VWF:Ag and its activity as measured using
Subtype 2A von Willebrand Disease. Ten percent to the VWF:RCo assay.
20% of VWD patients have subtype 2A, which arises from well-
characterized autosomal dominant point mutations in the A2 Subtype 2N von Willebrand Disease (Normandy
structural domain of the VWF molecule. These mutations Variant or Autosomal Hemophilia). A rare autosomal
render VWF more susceptible to proteolysis, which leads to a VWF missense mutation impairs its factor VIII binding site.
predominance of small-molecular-weight multimers in the This condition results in factor VIII deficiency despite normal
plasma. The smaller multimers support less platelet adhesion VWF:Ag concentration and activity and a normal multimeric
activity than the normal high- or intermediate-molecular- pattern. The disorder is also known as autosomal hemophilia
weight multimers. Patients with subtype 2A VWD have normal because its clinical symptoms are indistinguishable from the
or slightly reduced VWF:Ag levels with markedly reduced VWF symptoms of hemophilia except that it affects both males and
activity as a result of the loss of the high-molecular-weight and females. Subtype 2N is suspected when a girl or woman is
intermediate-molecular-weight multimers essential for platelet diagnosed with hemophilia after soft tissue bleeding symp-
adhesion.63 toms. In boys or men, subtype 2N is suspected when a patient
misdiagnosed with hemophilia A fails to respond to factor VIII
Subtype 2B von Willebrand Disease. In subtype 2B concentrate therapy. The poor response occurs because the
VWD, rare mutations within the A1 domain raise the affinity factor has a plasma half-life of mere minutes when it cannot
of VWF for platelet glycoprotein Ib/V/IX, its customary be bound by VWF. The diagnosis of VWD subtype 2N is made
binding site; these are thus gain of function mutations. Large by a molecular assay that detects the specific mutation respon-
VWF multimers spontaneously bind resting platelets and sible for the abnormal factor VIII binding to VWF.
are unavailable for normal platelet adhesion. Consequently,
the electrophoretic multimer pattern is characterized by Type 3 von Willebrand Disease. Autosomal recessive
lack of high-molecular-weight multimers but presence of VWF gene translation or deletion mutations produce severe
CHAPTER 41 Hemorrhagic Coagulation Disorders 657

TABLE 41-4 Laboratory Detection and Classification of von Willebrand Disease: Typical Results*
Laboratory Test Type 1 Type 2A Type 2B Type 3
VWF activity Low Low Low Very low
VWF antigen Low Normal to slightly decreased Normal to slightly decreased Very low
VWF activity to VWF >0.5 <0.5 <0.5 N/A
antigen ratio
Platelet count Normal Normal Decreased Normal
PTT Normal to slightly prolonged Normal Normal Prolonged
RIPA Decreased Decreased Increased Absent
Factor VIII activity Mildly low Normal Normal <5%
VWF multimers Normal pattern Large and intermediate forms absent Large forms absent All forms absent

*Expected results are given, but results vary over time and within affected kindreds.
N/A, Not applicable; PTT, partial thromboplastin time; RIPA, ristocetin-induced platelet aggregometry with 1mg/mL ristocetin; VWF, von Willebrand factor.

mucocutaneous and anatomic hemorrhage in compound het- VWF multimer analysis by sodium dodecyl sulfatepolyacrylamide
erozygotes or, in consanguinity, homozygotes. In this rare dis- gel electrophoresis further differentiates between VWD subtypes
order, VWF is absent or nearly absent from plasma. Factor VIII 2A and 2B. Although both 2A and 2B lack high-molecular-
is also proportionally diminished or absent, and primary and weight multimers, intermediate multimers are present in the
secondary hemostasis is impaired. electrophoretic pattern of subtype 2B VWD samples but are
absent from the pattern of a patient with subtype 2A VWD.
Laboratory Detection and Classification of Multimer analysis is technically demanding and is performed
von Willebrand Disease mainly in reference laboratories for differentiation of VWD
Definitive diagnosis of VWD depends on the combination of subtypes. Table 41-4 lists the expected test results for all VWD
a personal and family history of mucocutaneous bleeding and types and subtypes.
the laboratory demonstration of decreased VWF activity. A CBC Because acquired VWD may mimic any one of the VWD
is necessary to rule out thrombocytopenia as the cause of types and subtypes, results of the laboratory workup do not
mucocutaneous bleeding, and PT and PTT, which assess the help to differentiate it from the inherited condition. Review of
coagulation system, are part of the initial VWD workup. No the clinical history with emphasis on age at onset of bleeding,
longer recommended, according to the 2009 NLHBI VWD comorbid conditions, and family history may signal acquired
guidelines, are the bleeding time test and the PFA-100 or other disease rather than the congenital form.
automated functional platelet assays. These traditional screen-
ing tests generate conflicting sensitivity and specificity data. Pitfalls in the Diagnosis of von Willebrand Disease
The standard VWD test panel includes three assays: a quan- VWF activity is influenced by varying genetic penetrance, ABO
titative VWF test (VWF:Ag assay) employing an enzyme immu- blood group, inflammation, hormones, age, and physical
noassay or automated latex immunoassay technique, such as stress.64 Increased estrogen levels during the second and third
the Liatest (Diagnostica Stago, Asnires-sur-Seine, France); the trimesters of pregnancy nearly normalize plasma VWF activity
VWF activity test, which determines the factors ability to bind even in women with moderate VWF deficiency. However, VWF
to platelets, also known as the VWF:RCo assay; and a factor function and concentration decrease rapidly after delivery,
VIII activity assay. The VWF:RCo assay employs preserved which may lead to acute postpartum hemorrhage, for which
reagent platelets. The agonist ristocetin supports platelet agglu- the obstetrician is watchful. Oral contraceptive use and
tination in the presence of VWF. hormone replacement therapy also raise VWF activity, and
When the ratio of the VWF:RCo assay value to the VWF:Ag activity waxes and wanes with the menstrual cycle.
concentration is less than 0.5, 0.6, or 0.7 (ratio cutoff is gener- VWF activity rises substantially in acute inflammation such
ated from reference interval studies by local laboratories), qual- as occurs postoperatively, subsequent to trauma, or during an
itative or type 2 VWD is inferred. Additional tests are needed to infection. Physical stress such as cold, exertion, or childrens
confirm type 2 VWD and to differentiate subtype 2A from crying or struggling during venipuncture causes VWF activity to
subtype 2B. Low-dose ristocetin-induced platelet aggregometry rise. VWF activity rises when the tourniquet remains tied for
(RIPA), also called the ristocetin response curve, identifies subtype more than 1 minute prior to venipuncture and drops if the
2B. The low-dose RIPA test is performed on platelet-rich plasma. specimen is stored in the refrigerator prior to testing. VWD
In subtype 2B the patients platelets, because they are coated patients experience fluctuation in disease severity over time,
with abnormal VWF multimers, agglutinate in response to less and the clinical manifestations of the disease vary from person
than 0.5mg/mL ristocetin; sometimes they even agglutinate in to person within kindreds, despite the assumption that every-
response to 0.1mg/mL ristocetin. In comparison, normal plate- one in the family possesses the same mutation. When the
lets or platelets from a patient with subtype 2A agglutinate only clinical presentation suggests VWD, the VWD laboratory assay
at ristocetin concentrations greater than 0.5mg/mL. panel should be repeated until the results are conclusive.65
658 PART VIII Hemostasis and Thrombosis

Adding to VWD diagnostic confusion is concern over the TABLE 41-5 Von Willebrand Factor (VWF) Reference
variability in the results of the VWF:RCo assay, which is based Intervals by Blood Group
on platelet aggregometry but which is performed using a
Blood Group Reference Interval
variety of instruments and methods, including at least one
automated device, the BCS XP system with von Willebrand O 36-157%
reagent (Siemens Healthcare Diagnostics, Inc., Deerfield, Ill.). A 48-234%
Ristocetin avidity varies from lot to lot. In the United States, B 57-241%
proficiency surveys consistently reveal VWF:RCo assays to have AB 64-238%
an unimpressive interlaboratory coefficient of variation of 30% Population based: Low VWF <50%
and a least detectable activity range of 6% to 12%.66 Population based: von Willebrand disease <30%
Concern over the poor reproducibility of results of the VWF
activity assay has led to attempts at developing alternative
assays, the most promising of which is the VWF collagen binding are challenged with VWD managementfor example, nurses,
(VWF:CB) assay. VWF:CB employs type III collagen as its pharmacists, emergency department and primary care physi-
solid-phase target antigen. Developed in 1990, the VWF:CB cians, internists, surgeons, gynecologists, and hematologists.
assay produces results that more closely match those of the Laboratory professionals ensure that those who manage VWD
VWF:RCo assay than of VWF:Ag assays, because collagen type patients are aware of the effects of ABO group, estrogen levels,
III binds predominantly large VWF multimers; however, the inflammation, and physical stress on VWF activity, and that
target collagen composition needs to be standardized before they understand the strengths and limitations of VWF labora-
this assay achieves standard screening status.67 The VWF:CB tory assay panels, the proper interpretation of assay results, and
assay has better precision than the VWF:RCo assay.68 the availability of follow-up and confirmatory assays.
Many reference laboratories add the VWF:CB assay to their
initial VWD screening profile. They argue that the VWF:CB Treatment of von Willebrand Disease
assay detects abnormalities of VWF collagen binding, whereas Mild bleeding may resolve with the use of local measures,
the VWF:RCo assay detects abnormalities of VWF platelet such as limb elevation, pressure, and application of ice packs
binding and cite instances in which the VWF:CB value (the athletes acronym is RICE for rest, ice, compression, and
was abnormal when the VWF:RCo value was normal, and elevation).69 Moderate bleeding may respond to estrogen
vice-versa. and desmopressin acetate, which trigger the release of VWF from
In an effort to more closely reflect clinical reality and reduce storage organelles. Therapeutic dosages are monitored when
false-positive type 1 VWD diagnoses, the 2009 NHLBI VWD necessary using serial VWF:Ag concentration assays. Desmo-
guidelines have coined the nondisease description low VWF pressin acetate (1-desamino-8-D arginine vasopressin) is an
for the condition in which VWF activity and antigen concentra- antidiuretic hormone analogue used to control incontinence
tions are between 30% and 50% of normal, the ratio of in diabetes mellitus and bedwetting; release of VWF from
VWF:RCo to VWF:Ag is greater than 0.5, and factor VIII activ- storage organelles is a side-effect. Desmopressin acetate in its
ity is greater than 50% of normal. This suggested category oral form, DDAVP, or nasal spray form, Stimate (both from
reflects the difficulty in distinguishing mucocutaneous bleed- CSL Behring, King of Prussia, Pa.), is consistently effective for
ing from self-reported easy bruising and recognizes that low type 1 VWD and generally useful for subtype 2A. It is contra-
VWF activity and bleeding often associate coincidentally. To indicated for subtype 2B, however, because it causes the release
make a definite type 1 VWD diagnosis, 30% of normal VWF of abnormal VWF with increased affinity for platelet receptors,
activity is used as the cutoff. Molecular confirmation of type 1 which may intensify thrombocytopenia and lead to increased
VWD, currently but a dream because of the plethora of con- platelet activation and thrombosis. Because of its antidiuretic
tributing mutations, may soon become routine as molecular property, repeated doses may lead to hyponatremia (low serum
microchip technology meets the challenge. sodium). For this reason, it is necessary to monitor and regu-
VWF activity varies by ABO blood group, and expert panels late electrolytes during desmopressin acetate therapy.
have in the past recommended that laboratory directors main- -Aminocaproic acid (EACA; Amicar) or tranexamic acid
tain separate reference intervals for each group, as provided in (Cyklokapron) inhibits fibrinolysis and may help control
Table 41-5. The 2009 NHLBI VWD guidelines have recom- bleeding when used alone or in conjunction with desmopres-
mended against this practice, noting that despite the ABO sin acetate. Therapy using nonbiologic preparations is pre-
blood grouping and associated reference ranges, the major ferred over human plasmaderived biologic therapy, because
determinant of bleeding risk is low VWF. Therefore, VWF test nonbiologicals eliminate the risk of viral disease transmission
result reference intervals are now population based rather than and circumvent religious objections to receipt of human blood
ABO stratified. The internationally generalized cutoff of 50% products.
for low VWF and 30% for VWD is clinically reasonable, For treatment of severe VWD (type 3) and subtype 2B, three
although laboratory directors may choose to validate and commercially prepared human plasmaderived high-purity
adjust this cutoff in internal reference interval studies. preparations are available that provide a mixture of VWF and
In a well-managed tertiary care facility, laboratory scientists coagulation factor VIII. These are Humate-P, Alphanate, and
and physicians communicate regularly with medical staff who Wilate (FDA-cleared December 2009).70 The calculation of the
CHAPTER 41 Hemorrhagic Coagulation Disorders 659

TABLE 41-6 Therapeutic Strategies in von Willebrand quantitative disorders in which the factor VIII coagulant activ-
Disease ity and antigen concentration are in agreement, but in rare
cases low activity is seen despite normal antigen concentra-
Type Primary Approach Other Options
tion.73 The latter cases are due to qualitative or structural factor
1 Estrogen, DDAVP, EACA Factor VIII/VWF concentrate VIII abnormalities traditionally known as cross-reacting material
2A Estrogen, DDAVP, EACA Factor VIII/VWF concentrate positive disorders.74
2B Factor VIII/VWF concentrate EACA Male hemizygotes, whose sole X chromosome contains a
2N Factor VIII/VWF concentrate EACA factor VIII gene mutation, experience anatomic bleeding, but
3 Factor VIII/VWF concentrate Platelet transfusions female heterozygotes, who are carriers, do not. When a female
carrier has children with an unaffected man the chances of
DDAVP, Desmopressin acetate; EACA, -aminocaproic acid; VWF, von Willebrand hemophilia A inheritance can be described as follows: 25%
factor.
chance of a normal daughter, 25% chance of a carrier daughter,
25% chance of a normal son, and 25% chance of a hemophilic
proper dosage follows principles identical to those used for son. All sons of men with hemophilia A and noncarrier women
treatment of hemophilia A provided in the next section. Labo- are normal, whereas all daughters are obligate carriers of the
ratory monitoring by the VWF:Ag assay is essential to deter- disease.75 In addition, approximately 30% of newly diagnosed
mine if the given amount produced the target level of VWF and cases arise as a result of spontaneous germline mutations in
to follow its degradation between doses. individuals who have no family history of hemophilia A.76
Recombinant and affinity-purified factor VIII preparations Rarely, the symptoms of hemophilia A may be seen in females.
contain no VWF and cannot be used to treat VWD. Cryopre- This phenomenon could be due to true homozygosity or
cipitate and FP are less desirable alternatives because of the risk double heterozygosity, such as in the female offspring of a
of virus transmission, and the necessary FP volume per dose hemophilic father and a carrier mother. Other possibilities
may cause TACO. Therapy for bleeding secondary to acquired include a spontaneous germline mutation in the otherwise
VWD follows the same principles as delineated previously plus normal allele of a heterozygous female or a disproportional
treatment of the primary disease, if applicable. Therapeutic inactivation of the X chromosome with the normal gene,
recommendations for VWD are summarized in Table 41-6. termed extreme lyonization. Finally, VWD of the Normandy
subtype may present as mild hemophilia A in males and
Hemophilia A females.
The hemophilias are congenital single-factor deficiencies
marked by anatomic soft tissue bleeding. Second to VWD in Clinical Manifestations of Hemophilia A
prevalence among congenital bleeding disorders, hemophilias Hemophilia A causes anatomic bleeds with deep muscle and
occur in 1 in 10,000 individuals. Of those affected, 85% are joint hemorrhages; hematomas; wound oozing after trauma or
deficient in factor VIII, 14% are deficient in factor IX, and surgery; and bleeding into the central nervous system, perito-
1% are deficient in factor XI or one of the other coagulation neum, gastrointestinal tract, and kidneys. Acute joint bleeds
factors, such as factor II (prothrombin), V, VII, X, or XIII. Con- (hemarthroses) are exquisitely painful and cause temporary
genital deficiency of factor VIII is called classic hemophilia or immobilization. Chronic joint bleeds cause inflammation and
hemophilia A.71 eventual permanent loss of mobility, whereas bleeding into
muscles may cause nerve compression injury, with first tempo-
Factor VIII Deficiency rary, then lasting disability. Cranial bleeds lead to severe, debil-
Factor VIII is a two-chain, 285,000-D protein translated from itating, and durable neurologic symptoms, such as loss of
the X chromosome. When the coagulation cascade is activated, memory, paralysis, seizures, and coma, and may be rapidly
thrombin cleaves plasma factor VIII and releases a large poly- fatal. Bleeding may begin immediately after a triggering event
peptide called the B domain that dissociates from the molecule. or may become manifest after a delay of several hours. Some
This leaves behind a calcium-dependent heterodimer that bleeding seems to be spontaneous.
detaches from its VWF carrier molecule to bind phospholipid The diagnosis of hemophilia A begins with laboratory
and factor IXa. The VIIIa/IXa complex, sometimes called tenase, testing after the birth of an infant to a mother who has a family
cleaves and activates coagulation factor X at a rate 10,000 times history of hemophilia. In the absence of a family history, abnor-
faster than free IXa can cleave factor X. Consequently, factor mal bleeding in the neonatal period, which may appear as easy
VIII deficiency significantly slows the coagulation pathways bruising, bleeding from the umbilical stump, postcircumcision
production of thrombin and leads to hemorrhage. In vitro, bleeding, hematuria, or intracranial bleeding, is considered
factor VIII (and factor V) is labile and deteriorates at about 5% suspicious for hemophilia. Severe hemophilia usually is diag-
an hour at room temperature. nosed in the first year of life, whereas mild hemophilia may
not become apparent until a triggering event such as trauma,
Hemophilia A Genetics surgery, or dental extraction occurs in late childhood, adoles-
The gene for factor VIII spans 186kb of the X chromosome cence, or adulthood.
and is the site of various deletions, stop codons, and nonsense The laboratory diagnosis of coagulopathies in the newborn
and missense mutations.72 Most of these mutations result in or older infant is complicated by the requirement for an
660 PART VIII Hemostasis and Thrombosis

unhemolyzed specimen of at least 2mL in volume from tiny TABLE 41-7 Results of Clot-Based Screening Assays in
veins and by the typically low newborn levels of some coagula- Congenital Single-Factor Deficiencies*
tion factors. The PT and PTT are expected to be prolonged
Deficient
because of physiologically low levels of factors II (prothrom-
Factor PT PTT TCT Reflex Test
bin), VII, IX, and X even in full-term infants. Factor VIII levels
are similar to the levels in adults, even in infants who are born Fibrinogen Prolonged Prolonged Prolonged Fibrinogen assay
prematurely, which allows skillful laboratory staff to provide Prothrombin Prolonged Prolonged Normal Prothrombin, V, VII,
the correct diagnosis of hemophilia A using the factor VIII and X assays
activity assay. V Prolonged Prolonged Normal Prothrombin, V, VII,
The severity of hemophilia A symptoms is inversely propor- and X assays
tional to factor VIII activity. Hematologists classify an activity VII Prolonged Normal Normal VII assay
level of less than 1% as severe, associated with spontaneous or VIII Normal Prolonged Normal VIII, IX, and XI assays
exaggerated bleeding in the neonatal period. Activity levels of IX Normal Prolonged Normal VIII, IX, and XI assays
1% to 5% are seen in moderate hemophilia, which is usually X Prolonged Prolonged Normal Prothrombin, V, VII,
diagnosed in early childhood after symptoms become appar- and X assays
ent. In mild hemophilia, with activity levels of 5% to 20%, XI Normal Prolonged Normal VIII, IX, and XI assays
hemorrhage follows significant trauma and becomes a risk XIII Normal Normal Normal Factor XIII
factor mainly in surgery or dental extractions, and patients may quantitative assay
go for long periods without symptoms.
PT, Prothrombin time; PTT, partial thromboplastin time; TCT, thrombin clotting time.
*Results are valid when no anticoagulants are in use.
Hemophilia A Complications
As a result of frequent bleeds, hemophilic patients often
have debilitating and progressive musculoskeletal lesions and
deformities, and neurologic deficiencies after intracranial physiologic variables that affect factor VIII activity and VWF:Ag
hemorrhage. In addition, other effects of chronic diseases, assays, such as estrogen levels, inflammation, stress, and
such as limited productivity, low self-esteem, poverty, drug exercise.
dependency, and depression, are common problems. Before The laboratory scientist establishes a reference interval for
the advent of sterilized and recombinant factor concentrates, the VIII:VWF ratio using plasmas from 30 or more normal
chronic hepatitis often resulted from repeated exposure to women. If the ratio of the individual being tested is below the
blood products. Tragically, 70% of hemophilics treated before lower limit of the interval, she is likely to be a carrier. These
1984 are human immunodeficiency virus (HIV) positive or results may be influenced by extreme lyonization, variation in
have died from acquired immunodeficiency syndrome.77 VWF production, and analytical variables; consequently, if
carrier status is suspected and the VIII:VWF ratio is greater than
Laboratory Diagnosis of Hemophilia A the lower limit of the reference range, genetic testing may be
The laboratory workup for a suspected congenital single coagu- necessary to detect one of the many polymorphisms associated
lation factor deficiency starts with the PT, PTT, and thrombin with factor VIII deficiency.
time and continues with factor assays based on the results of
the screening tests. Before the physician initiates laboratory Hemophilia A Treatment
testing, however, a clear history of the patients hemorrhagic The goal of on-demand hemophilia A treatment is to raise the
symptoms must be recorded. In hemophilia A, the PT and patients factor VIII activity to hemostatic levels whenever he
thrombin time are normal, and the PTT is prolonged, provided experiences or suspects a bleeding episode or anticipates a
that the PTT reagent is sensitive to factor VIII deficiencies at hemostatic challenge such as a surgical procedure. The target
or less than the 30% plasma level. Table 41-7 lists the activity level depends on the nature of the bleeding, but it is
expected results for each clot-based screening assay for any seldom necessary to reach activity greater than 75%. Target
single-factor deficiency associated with bleeding, including activity should be maintained until the threat is resolved. In
deficiencies of fibrinogen and factors II (prothrombin), V, VII, the case of a bleed into soft tissue or a body cavity, the sooner
VIII, IX, X, and XI. Deficiencies of contact factors (factor XII, the target factor level is reached, the less painful is the episode,
high-molecular-weight kininogen, and prekallikrein) have no and the less likely is the patient to experience inflammation or
relationship to bleeding, and these factors do not appear in nerve damage. Because factor VIII has a half-life of 8 to 12
Table 41-7. hours, twice-a-day infusions are required.
With the availability of abundant recombinant factor VIII
Hemophilia A Carrier Detection concentrate, many hemophilic patients maintain themselves
Approximately 90% of female carriers of hemophilia A are on a steady prophylactic dosage designed to constantly keep
detected using the ratio of factor VIII activity to VWF:Ag value their factor VIII activity at hemostatic levels.78 Although it is
(VIII:VWF). This ratio is effective because VWF production is initially more expensive, the prophylactic approach saves
unaffected by factor VIII deficiency. Using a ratio, rather than downstream resources by ameliorating the adverse effects of
a factor VIII assay value alone, normalizes for some of the repeated hemorrhages and their long-term consequences.79
CHAPTER 41 Hemorrhagic Coagulation Disorders 661
Many hemophilic patients factor VIII activity rises upon VIII activity fails to rise to the target level after appropriate
administration of desmopressin acetate in the form of DDAVP concentrate administration. Most factor VIII inhibitors are
or Stimate (nasal formulation), alone or in combination with immunoglobulin G4, noncomplement-fixing, warm-reacting
an antifibrinolytic such as Amicar or Cyklokapron. When des- antibodies. It is impossible to predict which patients are likely
mopressin acetate treatment proves ineffective, intravenous to develop inhibitors based on genetics, demographics, or the
factor VIII concentrates are the next option. High-purity syn- type of concentrate used.85
thetic factor VIII concentrates are produced from mammalian The first step in inhibitor detection is a factor VIII assay. If
cells using recombinant deoxyribonucleic acid (DNA) technol- the factor VIII activity exceeds 30%, no inhibitor is present. If
ogy or from human plasma using factor VIIIspecific mono- the level is less than 30%, the laboratory scientist proceeds
clonal antibodies and column separation.80,81 The human to mixing studies. When the test plasma from the bleeding
plasmaderived concentrates Alphanate, Humate-P, and Wilate patient has a prolonged PTT, it is mixed 1:1 with NP and
are prepared by chemical fractionation of human plasma and incubated 1 to 2 hours at 37 C, and the PTT of the mixture
contain VWF, fibrinogen, and noncoagulant proteins, in addi- is measured. If no inhibitor is present, the incubated mixture
tion to factor VIII. All plasma-derived concentrates undergo should produce a normal or near-normal PTT result. If an
viral inactivation steps, and since 1985, none has transmitted inhibitor is present, however, the factor VIII from the normal
lipid-envelope viruses such as HIV, hepatitis B virus, or hepa- plasma is partially neutralized, and the mixtures PTT remains
titis C virus. Plasma-derived factor VIII concentrates may trans- prolonged or uncorrected, presumptive evidence of the
mit nonlipid viruses such as parvovirus B19 and hepatitis A inhibitor.
virus, however.82 Recombinant products may use human If mixing studies and the clinical picture suggest presence
albumin in the manufacturing process, which introduces the of a factor VIII inhibitor, a Bethesda assay is used to quantitate
theoretical risk of Creutzfeldt-Jakob disease transmission; the inhibitor. NP with 100% factor activity is mixed at increas-
however, manufacturers more recently have developed prod- ing dilutions in a series of tubes with the full-strength patient
ucts free of all human protein, such as Bioclate (Pfizer, New plasma. Factor VIII assays are performed on each mixture. The
York, N.Y.).83 operator then compares the results of the various dilutions and
When hematologists treat a hemophilic patient, they base expresses the titer as Bethesda units. One Bethesda unit is the
their factor VIII concentrate dosage calculations on the defini- reciprocal of the dilution that caused neutralization of 50% of
tion of a unit of factor VIII activity, which is taken as the the factor VIII from the NP. The same assay is employed
amount present in 1mL of normal plasma and is synonymous to measure factor VIII inhibitors in acquired hemophilia.
with 100% activity. They further calculate the desired increase Although the complex kinetics of acquired autoantibodies
after factor VIII concentrate infusion by subtracting the patients diminishes the accuracy of the results, this method is adequate
preinfusion factor activity from the target activity level. The to monitor therapy.
desired increase is multiplied by the patients plasma volume Hemophilia patients with inhibitors may be low or high
to compute the dosage. The patients plasma volume may be responders. Low responders have titers of 5 Bethesda units or
estimated from his or her blood volume and hematocrit.84 The less, and their titers do not increase significantly after factor
blood volume is approximately 65mL/kg of body weight, and VIII administration. High responders have inhibitor titers that
the plasma volume is the plasmacrit (100% hematocrit %) exceed 5 Bethesda units; their titer increases anamnestically
as in the following formulas: after therapy. Each laboratory may choose to maintain a data-
base of hemophilia patients who have inhibitors, because pre-
Plasma volume = weight in kilograms 65 mL kg vious titers often predict future inhibitor behavior.
(1 hematocrit )
Factor VIII concentrate dose = plasma volume Treatment of Hemophilia A in Patients
( target factor VIII level initial factor VIII level) with Inhibitors
Every hemophilic patient with an inhibitor needs an individu-
This formula is also used in the treatment of VWD. Overdos- alized treatment plan to control bleeding episodes. Low
ing seems to confer no thrombotic risk, but wastes resources. responders often experience cessation of bleeding upon admin-
Regardless of the dose administered, laboratory monitoring istration of large doses of factor VIII concentrate and may be
and close clinical observation are essential to prevent or halt so maintained. High responders may gain no benefit from
bleeding and its complications. Repeat dosing is done on an factor VIII concentrates and instead are treated with Autoplex
8- to 12-hour schedule reflecting the half-life of factor VIII. The T or FEIBA, which generates thrombin in the presence of factor
second administration of factor VIII uses half the concentration VIII inhibitors. FEIBA dosage should not exceed 200 units/kg
of the first dose. per day, distributed in two to four injections, because the acti-
vated factors may trigger DIC. NovoSeven also bypasses the
Hemophilia A and Factor VIII Inhibitors physiologic factor VIII requirement, because it promotes
Alloantibody inhibitors of factor VIII arise in response to treat- thrombin formation through the tissue factor pathway.86
ment in 30% of patients with severe hemophilia and 3% of The discussions in this chapter of ACOTS and acquired hemo-
those with moderate hemophilia. The presence of an inhibitor philia provide additional detail on FEIBA and NovoSeven
is suspected when bleeding persists or when the plasma factor dosages.
662 PART VIII Hemostasis and Thrombosis

Vascular injury dose is doubled to compensate for factor IX distribution into


TF
the extravascular space. Repeat doses of factor IX are given
every 24 hours, which reflects the half-life of the factor. The
VIIa IXa XIa second and subsequent doses, if needed, are half the initial
dose, provided that factor assays determine that the target level
of factor IX was achieved.
VIIIa
Inhibitors to factor IX arise in only 3% of hemophilia B
Xa patients. Antifactor IX alloantibodies react avidly with factor
Va
IX and may be detected using the Bethesda assay. Bleeding in
patients with inhibitors is treated as in patients with factor VIII
inhibitors using Autoplex T, FEIBA, or NovoSeven.
IIa

Hemophilia C (Rosenthal Syndrome,


Factor XI Deficiency)
Fibrin
Factor XI deficiency is an autosomal dominant hemophilia
polymer
with mild to moderate bleeding symptoms. More than half of
XIIIa
the cases have been described in Ashkenazi Jews, but individu-
als of any ethnic group may be affected. The frequency and
Cross-linked severity of bleeding episodes do not correlate with factor XI
fibrin levels, and laboratory monitoring of treatment serves little
purpose after the diagnosis is established. The physician treats
Thrombus hemophilia C with frequent infusions of FP during times of
Figure 41-5 Simplified coagulation cascade mechanism showing posi- hemostatic challenge.88 In the laboratory, the PTT is prolonged
tions of all factors whose absence may cause hemorrhage. TF, Tissue factor.
and the PT is normal.

Other Congenital Single-Factor Deficiencies


Hemophilia B (Factor IX Deficiency) The remaining congenital single-factor deficiencies listed in
Hemophilia B, also called Christmas disease, represents approxi- Table 41-8 are rare, are caused by autosomal recessive muta-
mately 14% of hemophilia cases in the United States, although tions, and are often associated with consanguinity. The PT,
its incidence in India nearly equals that of hemophilia A.87 PTT, and thrombin time may be employed to distinguish
Hemophilia B is caused by deficiency of factor IX, one of the among these disorders, as shown in Table 41-7. In addition,
vitamin Kdependent serine proteases. Factor IX is a substrate immunoassays may be performed to distinguish among the
for both factors XIa and VIIa because it is cleaved by either to more prevalent quantitative and the less prevalent qualitative
form dimeric factor IXa (Figure 41-5). Subsequently, factor IXa abnormalities. In qualitative disorders, often called dysprotein-
complexes with factor VIIIa to cleave and activate its substrate, emias, the ratio of factor activity to antigen is less than 0.7. The
factor X. Factor IX deficiency reduces thrombin production and bleeding symptoms in the dysproteinemias may be more
causes soft tissue bleeding that is indistinguishable from that severe than in quantitative deficiencies, but the risk of inhibitor
in hemophilia A. It also is a sex-linked, markedly heteroge- formation is theoretically lower. The clot-based measurement
neous disorder involving more than 220 separate mutations of factors II (prothrombin) and X may be supplemented or
resulting in a range of mild to severe bleeding manifestations. replaced by more reproducible chromogenic substrate assays.
Determination of carrier status is less successful in hemophilia Fibrinogen is usually measured using the Clauss assay, a modi-
B than in hemophilia A because of the large number of factor fication of the thrombin time, but may be measured by turbi-
IX mutations and the lack of a linked molecule such as VWF dimetry or immunoassay.
that can be used as a normalization index. DNA analysis occa- Because platelets transport about 20% of circulating factor
sionally may be used to establish carrier status when hemo- V, the platelet function in factor V deficiency may be diminished,
philia B has been diagnosed and its specific mutation identified which is reflected in a prolonged bleeding time but normal
in a relative. platelet aggregation.89 The PT and PTT are prolonged. Because
The laboratory is essential to the diagnosis of hemophilia of the concentration of factor V in platelet -granules, normal
B. The PTT typically is prolonged, whereas the PT is normal. If platelet concentrate is an effective form of therapy for factor V
the clinical symptoms suggest hemophilia B, the factor IX assay deficiency. A combined factor V and VIII deficiency may be
should be performed even if PTT is within the reference range, caused by a genetic defect traced to chromosome 18 that affects
because the PTT reagent may be insensitive to mild factor IX transport of both factors by a common protein in the Golgi
deficiency. apparatus.90
Recombinant or column-purified plasma-derived factor IX Factor VII deficiency causes moderate to severe anatomic
concentrates are used to treat hemophilia B. Dosing is calcu- hemorrhage. The bleeding does not necessarily reflect the factor
lated the same way as for factor VIII concentrates in hemo- VII activity level. The half-life of factor VII is approximately 6
philia A and VWD, with the exception that the calculated initial hours, which affects the frequency of therapy. NovoSeven at
CHAPTER 41 Hemorrhagic Coagulation Disorders 663

TABLE 41-8 Rare Congenital Single-Factor Deficiencies


Deficiency Factor Levels Symptoms Therapy
Afibrinogenemia No measurable fibrinogen Severe anatomic bleeding CRYO to raise to >100mg/dL
Hypofibrinogenemia Fibrinogen activity assay <100mg/dL Moderate systemic bleeding CRYO to raise to >100mg/dL
Dysfibrinogenemia Fibrinogen activity assay <100mg/dL Mild systemic bleeding CRYO to raise to >100mg/dL
Prothrombin deficiency Factor II <30% Mild systemic bleeding PCC or plasma to raise to 75%
Factor V deficiency Factor V <30% Mild systemic bleeding Plasma to raise to 75%
Factor VII deficiency Factor VII <30% Moderate to severe anatomic bleeding Activated factor VII, plasma
Factor X deficiency Factor X <30% Severe anatomic bleeding PCC or plasma to raise to 75%
Factor XI deficiency Factor XI <50% Anatomic bleeding Plasma to raise to 75%
Factor XIII deficiency Factor XIII <1% Moderate to severe systemic bleeding, Plasma or CRYO every 3wk
poor wound healing

CRYO, Cryoprecipitate; PCC, prothrombin complex concentrate.

30mcg/mL and prothrombin complex concentrate such as TABLE 41-9 Factor XIII Deficiency
Autoplex T preparations are effective and may provide a target
Type of Factor XIII
level of 10% to 30%. Many factor VII deficiencies are dyspro-
Deficiency Incidence Activity -Protein -Protein
teinemias.91 The PT, but not the PTT, is prolonged in factor VII
deficiency. Type I Rare Absent Absent Absent
Factor X deficiency causes moderate to severe anatomic hem- Type II Frequent Absent Normal Low
orrhage that may be treated with FP or prothrombin complex Type III Rare Low Absent Low
concentrate to produce therapeutic levels of 10% to 40%.92 The
half-life of factor X is 24 to 40 hours. Acquired factor X defi-
ciency has been described in amyloidosis, in paraproteinemia, chain is a binding and stabilizing portion. Factor XIII defi-
and in association with antifungal drug therapy. The hemor- ciency occurs in three forms related to the affected chain, as
rhagic symptoms are severe and life-threatening. The PT and shown in Table 41-9. Patients with factor XIII deficiency have
PTT are both prolonged in factor X deficiency. In the time- a normal PT, PTT, and thrombin time despite anatomic bleeds
honored Russell viper venom time test, which activates the coagu- and poor wound healing. They form weak (noncross-linked)
lation mechanism at the level of factor X, clotting time is clots that dissolve within 2 hours when suspended in a 5-M
prolonged in deficiencies of factors X and V, prothrombin, and urea solution, a traditional factor XIII screening assay.93 To
fibrinogen. The venom used is harvested from the Russell viper, confirm factor XIII deficiency, factor activity may be measured
the most dangerous snake in Asia. This test may be useful in accurately using a chromogenic substrate assay such as the
distinguishing a factor VII deficiency, which does not affect the Behring Behrichrom FXIII assay (Behring Diagnostics, King of
results, from deficiencies in the common pathway, although Prussia, Pa.).
specific factor assays are the standard approach. Finally, autosomally inherited deficiencies of the fibrino-
Plasma factor XIII is a tetramer of paired and mono- lytic regulatory proteins 2-antiplasmin and plasminogen acti-
mers. The intracellular form is a homodimer (two chains) vator inhibitor-1 have been reported to cause moderate to
and is stored in platelets, monocytes, placenta, prostate, and severe bleeding. Both are rare and may be diagnosed using
uterus. The chain contains the active enzyme site, and the chromogenic substrate assays.

SUMMARY
Hemorrhage can be classified as localized versus generalized, The hemophilias are congenital single-factor deficiencies that
acquired versus congenital, and anatomic soft tissue versus cause moderate to severe anatomic hemorrhage. The clinical
mucocutaneous. laboratory plays a key role in diagnosis, classification, and treat-
Acquired hemorrhagic disorders that are diagnosed in the hemo- ment monitoring in hemophilia.
stasis laboratory include thrombocytopenia of various etiologies,
ACOTS, liver and renal disease, vitamin K deficiency, scurvy, Now that you have completed this chapter, go back and
acquired hemophilia or VWD, and DIC. read again the case study at the beginning and respond
VWD is the most common congenital bleeding disorder, and the to the questions presented.
diagnosis and classification of the type and subtypes require a
series of clinical laboratory assays.
664 PART VIII Hemostasis and Thrombosis

R E V I E W Q UESTIONS
1. What is the most common acquired bleeding disorder? 7. If a patient has anatomic soft tissue bleeding and poor
a. ACOTS wound healing, but the PT, PTT, thrombin time, platelet
b. Vitamin K deficiency count, and platelet functional assay results are normal, a
c. Liver disease deficiency of what factor could exist?
d. VWD a. Fibrinogen
b. Prothrombin
2. Which is a typical form of anatomic bleeding? c. Factor XII
a. Epistaxis d. Factor XIII
b. Menorrhagia
c. Hematemesis 8. What therapy may be used for a hemophilic boy who
d. Soft tissue bleed is bleeding and who has a high titer of factor VIII
inhibitor?
3. To a deficiency of which factor is the PT most sensitive? a. FP
a. Prothrombin b. Cryoprecipitate
b. VII c. Factor VIII concentrate
c. VIII d. Recombinant activated coagulation factor VII
d. IX
9. What is the most prevalent form of VWD?
4. Which of the following conditions causes a prolonged a. Type 1
thrombin time? b. Type 2A
a. Prothrombin deficiency c. Type 2B
b. Antithrombin deficiency d. Type 3
c. Hypofibrinogenemia
d. Warfarin therapy 10. Which of the following assays can be used to distinguish
vitamin K deficiency from liver disease?
5. In what subtype of VWD is the RIPA test result positive a. Factor V assay
when ristocetin is used at a concentration of less than b. PT
0.5mg/mL? c. Factor VII assay
a. Subtype 2A d. Protein C assay
b. Subtype 2B
c. Subtype 2N 11. Mucocutaneous hemorrhage is typical of:
d. Type 3 a. Acquired hemorrhagic disorders
b. Localized hemorrhagic disorders
6. What is the typical treatment for vitamin K deficiency c. Defects in primary hemostasis
when the patient is bleeding? d. Defects in fibrinolysis
a. Vitamin K and FP
b. Vitamin K and platelet concentrate
c. Vitamin K and factor VIII concentrate
d. Vitamin K and prothrombin complex concentrate

REFERENCES
1. Kessler CM, Khokhar N, Liu M: A systematic approach to the hemorrhage. On behalf of the NovoSeven Trauma Study
bleeding patient: correlation of clinical symptoms and signs Group. Transfusion 49:240S-247S, 2009.
with laboratory testing. In Kitchens CS, Alving BM, Kessler 5. Duchesne JC, Holcomb JB: Damage control resuscitation:
CM, editors: Consultative hemostasis and thrombosis, ed 2, addressing trauma-induced coagulopathy. Br J Hosp Med
St Louis, 2007, Saunders, pp 17-30. (Lond) 70:22-25, 2009.
2. Zumber M, Kitchens CS: Purpura and other hematovascular 6. Shakur H, Roberts I, Bautista R, et al: CRASH-2 Trial Collabo-
lesions. In Kitchens CS, Alving BM, Kessler CM, editors: rators: Effects of tranexamic acid on death, vascular occlusive
Consultative hemostasis and thrombosis, ed 2, St Louis, 2007, events, and blood transfusion in trauma patients with
Saunders, pp 159-182. significant haemorrhage (CRASH-2): a randomised, placebo-
3. Miller R, Beeton K, Goldman E, et al: Counseling guidelines controlled trial. Lancet 376(9734):23-32, 2010.
for managing musculoskeletal problems in haemophilia in 7. Hathaway WE, Goodnight SH: Screening tests of hemostasis.
the 1990s. Haemophilia 3:9-13, 1977. In Hathaway WE, Goodnight SH, editors: Disorders of
4. Boffard KD, Choog PIT, Kloger Y, et al: The treatment of hemostasis and thrombosis, New York, 2001, McGraw-Hill,
bleeding is to stop the bleeding! Treatment of trauma-related pp 41-51.
CHAPTER 41 Hemorrhagic Coagulation Disorders 665
8. Weiss AE: Acquired coagulation disorders. In Corriveau 27. Horan JT, Francis CW: Fibrin degradation products, fibrin
DM, Fritsma GA, editors: Hemostasis and thrombosis in monomer and soluble fibrin in disseminated intravascular
the clinical laboratory, Philadelphia, 1988, JB Lippincott, coagulation. Semin Thromb Hemost 27:657-666, 2001.
pp 169-205. 28. Mammen EF: Disseminated intravascular coagulation (DIC).
9. US Centers for Disease Control and Prevention: Web-based Clin Lab Sci 13:239-245, 2000.
Injury Statistics Query and Reporting System. Available 29. Schwartz RS, Bauer KA, Rosenberg RD, et al: Clinical experi-
at: http://www.cdc.gov/injury/wisqars/index.html. Accessed ence with antithrombin III concentrate in treatment of con-
June 24, 2010. genital and acquired deficiency of antithrombin. Am J Med
10. Boffard KD, Choog PIT, Kloger Y, et al: The treatment of 87(suppl 3b):53S-60S, 1989.
bleeding is to stop the bleeding! Treatment of trauma-related 30. Dettenmeier P, Swindell B, Stroud M, et al: Role of activated
hemorrhage. On behalf of the NovoSeven Trauma Study protein C in the pathophysiology of severe sepsis. Am J Crit
Group. Transfusion 49:240S-247S, 2009. Care 12:518-524, 2003.
11. Duchesne JC, Holcomb JB: Damage control resuscitation: 31. Saito H: Alterations of hemostasis in renal disease. In
addressing trauma-induced coagulopathy. Br J Hosp Med Ratnoff OD, Forbes CD, editors: Disorders of hemostasis, ed 3,
(Lond) 70:22-25, 2009. Philadelphia, 1996, Saunders, pp 443-456.
12. McLaughlin DF, Niles SE, Salinas J, et al: A predictive model 32. Galbusera M, Remuzzi G, Boccardo P: Treatment of bleeding
for massive transfusion in combat casualty patients. J Trauma in dialysis patients. Semin Dial 22:279-286, 2009.
64:S57-S63, 2008. 33. Rabiner SF: Uremic bleeding. Prog Hemost Thromb 1:233-250,
13. Practice guidelines for perioperative blood transfusion and 1972.
adjuvant therapies: an updated report by the American 34. Kahan BD: Fifteen years of clinical studies and clinical prac-
Society of Anesthesiologists Task Force on Perioperative tice in renal transplantation: reviewing outcomes with de
Blood Transfusion and Adjuvant Therapies. Anesthesiology novo use of sirolimus in combination with cyclosporine.
105:198-208, 2006. Transplant Proc 40:S17-S20, 2008.
14. Kakaiya R, Aronson CA, Julleis J: Whole blood collection and 35. Leng SX, Elias JA: Interleukin-11. Int J Biochem Cell Biol
component procession. In Roback JD, Combs MR, Grossma 29:1059-1062, 1997.
BJ, et al, editors: Technical manual, ed 16, Bethesda Md, 2008, 36. Ambuhl PM, Wuthrich RP, Korte W, et al: Plasma hyper
AABB. coagulability in haemodialysis patients: impact of dialysis
15. Downes KA, Wilson E, Yomtovian R, et al: Serial measure- and anticoagulation. Nephrol Dial Transplant 12:2355-2364,
ment of clotting factors in thawed plasma stored for five days. 1997.
Transfusion 41:570 (letter), 2001. 37. Lind SE: The bleeding time does not predict surgical bleeding.
16. Borgman MA, Spinella PC, Perkins JG, et al: The ratio of Blood 77:2547-2552, 1991.
blood products transfused affects mortality in patients receiv- 38. Wadhwa M, Thorpe R: Haematopoietic growth factors and
ing massive transfusions in a combat support hospital. their therapeutic use. Thromb Haemost 99:863-873, 2008.
J Trauma 63:805-813, 2007. 39. Brunzel NA: Renal and metabolic disease. In Fundamentals
17. Levi M, Peters M, Buller HR: Efficacy and safety of recombi- of urine and body fluid analysis, ed 2, Philadelphia, 2002,
nant factor VIIa for treatment of severe bleeding: a systematic Saunders, pp 271-310.
review. Crit Care Med 33:883-890, 2005. 40. Kauffmann R, Veltkamp J, van Tilburg N, et al: Acquired
18. Tatoulis J, Theodore S, Meswani M, et al: Safe use of recom- antithrombin III deficiency and thrombosis in the nephrotic
binant activated factor VIIa for recalcitrant postoperative syndrome. Am J Med 298:562-571, 1978.
haemorrhage in cardiac surgery. Interact Cardiovasc Thorac 41. Bleyer WA, Hakami N, Shepard TH: The development of
Surg 9:459-462, 2009. hemostasis in the human fetus and newborn infant. J Pediatr
19. OConnell KA, Wood JJ, Wise RP, et al: Thromboembolic 79:838-853, 1971.
adverse events after use of recombinant human coagulation 42. Zipursky A, Desa D, Hsu E, et al: Clinical and laboratory
factor VIIa. JAMA 295:293-298, 2006. diagnosis of hemostatic disorders in newborn infants. Am J
20. Robert A, Chazouilleres O: Prothrombin time in liver failure: Pediatr Hematol Oncol 1:217-226, 1979.
time, ratio, activity percentage, or international normalized 43. Misra D, Bednar M, Cromwell C, et al: Manifestations of
ratio? Hepatology 24:1392-1394, 1996. superwarfarin ingestion: a plea to increase awareness. Am J
21. Senzolo M, Burroughs AK: Hemostatic alterations in liver Hematol 85:391-392, 2010.
disease and liver transplantation. In Kitchens CS, Alving BM, 44. Kershaw G, Jayakodi D, Dunkley S: Laboratory identification
Kessler CM, editors: Consultative hemostasis and thrombosis, of factor inhibitors: the perspective of a large tertiary hemo-
ed 2, St Louis, 2007, Saunders, pp 647-660. philia center. Semin Thromb Hemost 35:760-768, 2009.
22. Ratnoff OD: Hemostatic defects in liver and biliary tract 45. Green D: Spontaneous inhibitors to coagulation factors. Clin
disease and disorders of vitamin K metabolism. In Ratnoff Lab Haematol 22(suppl 1):21-25, 2000.
OD, Forbes CD, editors: Disorders of hemostasis, ed 3, 46. Biggs R, Austen DEG, Denson DWE, et al: The mode of action
Philadelphia, 1996, Saunders, pp 422-442. of antibodies which destroy factor VIII: II. Antibodies which
23. Lisman T, Leebeek FW, de Groot PG: Haemostatic abnor give complex concentration graphs. Br J Haematol 23:137-
malities in patients with liver disease. J Hepatol 37:280-287, 155, 1972.
2002. 47. Miesbach W, Matthias T, Scharrer I: Identification of throm-
24. Ferro D, Quintarelli C, Lattuada A, et al: High plasma levels bin antibodies in patients with antiphospholipid syndrome.
of von Willebrand factor as a marker of endothelial perturba- Ann N Y Acad Sci 1050:250-256, 2005.
tion in cirrhosis: relationship to endotoxemia. Hepatology 48. Krumdieck R, Shaw DR, Huang ST, et al: Hemorrhagic disor-
23:1377-1383, 1996. der due to an isoniazid-associated acquired factor XIII inhibi-
25. Hollestelle MJ, Geertzen HG, Straatsburg JH, et al: Factor VIII tor in a patient with Waldenstrms macroglobulinemia. Am
expression in liver disease. Thromb Haemost 91:267-275, J Med 90:639-645, 1991.
2004. 49. Shapiro SS, Hultin M: Acquired inhibitors to the blood coag-
26. Younger HM, Hadoke PW, Dillon JF, et al: Platelet function ulation factors. Semin Thromb Hemost 1:336-385, 1975.
in cirrhosis and the role of humoral factors. Eur J Gastroenterol 50. Israels SJ, Israels ED: Development of antibodies to bovine
Hepatol 9:982-992, 1997. and human factor V in two children after exposure to topical
666 PART VIII Hemostasis and Thrombosis

bovine thrombin. Am J Pediatr Hematol Oncol 16:249-254, Willebrand factor/factor VIII concentrate. Haemophilia
1994. 15:122-130, 2009.
51. Barker B, Altuntas F, Paranjape G, et al: Presurgical plasma 71. Boggio LN, Kessler CM: Hemophilia A and B. In Kitchens CS,
exchange is ineffective in correcting amyloid associated factor Alving BM, Kessler CM, editors: Consultative hemostasis and
X deficiency. J Clin Apheresis 19:208-210, 2004. thrombosis, ed 2, St Louis, 2007, Saunders, pp 45-60.
52. von Depka M: Immune tolerance therapy in patients with 72. DiMichelle DM: Hemophilia A (FVIII deficiency). In
acquired hemophilia. Hematology 9:245-257, 2004. Hathaway WE, Goodnight SH, editors: Disorders of hemosta-
53. Kumar S, Pruthri RK, Nichols WL: Acquired von Willebrand sis and thrombosis, ed 2, New York, 2001, McGraw-Hill,
disease. Mayo Clin Proc 77:181-187, 2002. pp 127-139.
54. Franchini M, Lippi G: Acquired von Willebrand syndrome: 73. Weiss AE: The hemophilias. In Corriveau DM, Fritsma GA,
an update. Am J Hematol 82:368-375, 2007. editors: Hemostasis and thrombosis in the clinical laboratory,
55. National Heart, Lung, and Blood Institute: The diagnosis, Philadelphia, 1988, JB Lippincott, pp 128-168.
evaluation, and management of von willebrand disease. NIH 74. Lusher JM, Warrier I: Hemophilia A. Hematol Oncol Clin North
Publication No. 08-5832, Bethesda, Md, 2009, US Department Am 6:1021-1033, 1992.
of Health and Human Services, National Institutes of Health. 75. Peake IR, Lillicrap DP, Boulyjenkov V, et al: Hemophilia:
56. James AH, Manco-Johnson MJ, Yawn BP, et al: Von strategies for carrier detection and prenatal diagnosis. Bull
Willebrand disease: key points from the 2008 National Heart, World Health Org 71:429-458, 1993.
Lung, and Blood Institute guidelines. Obstet Gynecol 114:674- 76. Rossetti LC, Radic CP, Larripa IB, et al: Genotyping the hemo-
678, 2009. philia inversion hotspot by use of inverse PCR. Clin Chem
57. Miller JL: von Willebrand disease. Hematol Oncol Clin North 51:1154-1158, 2005.
Am 4:107-123, 1990. 77. Evatt BL: The tragic history of AIDS in the hemophilia popu-
58. Ginsburg D, Sadler JE: von Willebrand disease: a database of lation, 1982-1984. J Thromb Haemost 4:2295-2301, 2006.
point mutations, insertions and deletions. For the Consor- 78. Gringeri A, Lundin V, von Mackensen S, et al: Primary and
tium on von Willebrand Factor Mutations and Polymor- secondary prophylaxis in children with haemophilia A
phisms, and the Subcommittee on von Willebrand Factor of reduces bleeding frequency and arthropathy development
the Scientific and Standardization Committee of the Interna- compared to on demand treatment; a 10-year, randomized
tional Society of Thrombosis and Haemostasis. Thromb clinical trial. J Thromb Haemost 7:780-786, 2009.
Haemost 69:177-184, 1993. 79. Schobess R, Kurnik K, Friedrichs F, et al: Effects of primary
59. Ginsburg D, Bowie EJW: Molecular genetics of von Wille- and secondary prophylaxis on the clinical expression of joint
brand disease. Blood 79:2507-2519, 1992. damage in children with severe haemophilia A. Results of a
60. Lee CA, Abdul-Kadir R: Von Willebrand disease and womens multicenter non-concurrent cohort study. Thromb Haemost
health. Semin Hematol 42:42-48, 2005. 99:71-76, 2008.
61. Rick ME: Von Willebrand disease. In Kitchens CS, Alving BM, 80. Bray GL, Gomperts ED, Courter S, et al: A multicenter study
Kessler CM, editors: Consultative hemostasis and thrombosis, of recombinant factor VIII (Recombinate): safety, efficacy,
ed 2, St Louis, 2007, Saunders, pp 97-110. and inhibitor risk in previously untreated patients with
62. Sadler JE, Rodeghiero F, on behalf of the ISTH SSC Subcom- hemophilia A. Blood 83:2428-2437, 1994.
mittee on von Willebrand Factor: Provisional criteria for the 81. Lusher JM, Arkin S, Abildgaard CF, et al: Recombinant factor
diagnosis of VWD type 1. J Thromb Haemost 3:775-777, 2005. VIII for the treatment of previously untreated patients with
63. Schneppenheim R, Budde U: Phenotypic and genotypic diag- hemophilia A. N Engl J Med 328:453-457, 1993.
nosis of von Willebrand disease: a 2004 update. Semin 82. Grner A: Pathogen safety of plasma-derived products
Hematol 42:15-28, 2005. Haemate P/Humate-P. Haemophilia Suppl 5:54-71, 2008.
64. Gill JC, Endres-Brooks J, Bauer PJ, et al: The effect of ABO 83. Kessler CM, Gill JC, White GC, et al: B-domain deleted
blood group on the diagnosis of von Willebrand disease. recombinant factor VIII preparations are bioequivalent to a
Blood 69:1691-1695, 1987. monoclonal antibody purified plasma-derived factor VIII
65. Favaloro EJ, Bonar R, Kershaw G, et al: Royal College of concentrate: a randomized, three-way crossover study. Hae-
Pathologists of Australasia Quality Assurance Program in mophilia 11:84-91, 2005.
Haematology: Laboratory diagnosis of von Willebrand disor- 84. Marques MB, Fritsma GA: Quick guide to coagulation testing,
der: use of multiple functional assays reduces diagnostic error ed 2, Washington, DC, 2009, American Association for Clini-
rates. Lab Hematol 11:91-97, 2005. cal Chemistry Press, p 57.
66. Hayes TE, Brandt JT, Chandler WL, et al: External peer review 85. McMillan EW, Shapiro SS, Whitehurst D, et al: The natural
quality assurance testing in von Willebrand disease: the history of factor VIII inhibitors in patients with hemophilia
recent experience of the United States College of American A: a national cooperative study: II. Observations on the initial
Pathologists proficiency testing program. Semin Thromb development of factor VIII inhibitors. Blood 71:344-348,
Hemost 32:499-504, 2006. 1988.
67. Favaloro EJ, Kershaw G, McLachlan AJ, et al: Time to think 86. Franchini M, Zaffanello M, Veneri D: Recombinant factor
outside the box? Proposals for a new approach to future VIIa: an update on its clinical use. Thromb Haemost 93:1027-
pharmacokinetic studies of von Willebrand factor concen- 1035, 2005.
trates in people with von Willebrand disease. Semin Thromb 87. Kessler CM, Mariani G: Clinical manifestations and therapy
Hemost 33:745-758, 2007. of the hemophilias. In Colman RW, Marder VJ, Clowes AW,
68. Favaloro EJ, Bonar R, Kershaw G, et al: Reducing errors in et al, editors: Hemostasis and thrombosis, ed 5, Philadelphia,
identification of von Willebrand disease: the experience of 2006, Lippincott Williams & Wilkins, pp 887-905.
the Royal College of Pathologists of Australasia quality assur- 88. Walsh PN, Gailani D: Factor XI. In Colman RW, Marder VJ,
ance program. Semin Thromb Hemost 32:505-513, 2006. Clowes AW, et al, editors: Hemostasis and thrombosis: basic
69. Rodeghiero F, Castaman G: Treatment of von Willebrand principles and clinical practice, ed 5, Philadelphia, 2006,
disease. Semin Hematol 42:29-35, 2005. Lippincott Williams & Wilkins, pp 221-235.
70. Berntorp E, Windyga J: European Wilate Study Group: Treat- 89. Miletich JP, Majerus DW, Majerus PW: Patients with congeni-
ment and prevention of acute bleedings in von Willebrand tal factor V deficiency have decreased factor Xa binding sites
diseaseefficacy and safety of Wilate, a new generation von on their platelets. J Clin Invest 62:824-831, 1978.
CHAPTER 41 Hemorrhagic Coagulation Disorders 667
90. Nichols WC, Seligsohn U, Zivelin A, et al: Linkage of com- 92. Fair DS, Edgington TS: Heterogeneity of hereditary and
bined factors V and VIII deficiency to chromosome 18q by acquired factor X deficiencies by combined immunochemical
homozygosity mapping. J Clin Invest 99:596-601, 1997. and functional analyses. Br J Haematol 59:235-248, 1985.
91. Chaing S, Clarke B, Sridhara S, et al: Severe factor VII defi- 93. Jenning I, Kitchen S, Woods TAL, et al: Problems relating to
ciency caused by mutations abolishing the cleavage site for the laboratory diagnosis of factor XIII deficiency: a UKNEQAS
activation and altering binding to tissue factor. Blood 83:3524- study. Thromb Haemost 1:2603-2608, 2003.
3535, 1994.
42 Thrombosis Risk Testing
George A. Fritsma*
OUTLINE OBJECTIVES
Developments in After completion of this chapter, the reader will be able to:
Thrombosis Risk Testing
Introduction to Thrombosis 1. Describe the prevalence of thrombotic disease in 9. Develop, carry out, and report the results of an anti-
Etiology of Thrombosis developed countries. thrombin testing protocol.
Prevalence of Thrombosis 2. Define thrombophilia. 10. List and employ tests of the protein C pathway,
Thrombosis Risk Factors 3. Distinguish between venous and arterial thrombosis. including protein C and protein S activity and con-
Acquired Thrombosis Risk 4. Differentiate among acquired thrombosis risk factors centration, activated protein C resistance, and the
Factors related to lifestyle and disease, and congenital risk factor V Leiden assay, to assess thrombotic risk.
Thrombosis Risk Factors factors, noting which can be assessed in the hemo- 11. Explain the molecular test for prothrombin G20210A
Associated with stasis laboratory. mutation and the relationship of this mutation to
Systemic Diseases 5. Discuss in relative terms, such as most commonly venous thrombosis, and interpret given test results.
Congenital Thrombosis
affected or uncommonly affected, the frequency 12. Describe the causes and pathophysiology of dis-
Risk Factors
with which various heritable risk factors occur in seminated intravascular coagulation (DIC).
Double Hit
Laboratory Evaluation of different ethnic groups. 13. List and perform the assays comprising a primary
Thrombophilia 6. Show how a hemostasis laboratory scientist may test profile for diagnosis and management of DIC in
Antiphospholipid Antibodies communicate with clinicians regarding the use of an acute care facility.
Activated Protein C thrombosis risk profiles and screening tests. 14. Discuss the value of quantitative D-dimer assays.
Resistance and Factor 7. List and employ the tests for predictors of arterial 15. Describe the efficacy of the assays used to
V Leiden Mutation thrombotic disease, such as high-sensitivity C- monitor localized thrombosis: prothrombin fragment
Prothrombin G20210A reactive protein, homocysteine, fibrinogen, lipopro- 1+2, fibrinopeptide A, and thrombin-antithrombin
Antithrombin tein (a), and factor assays, to assess thrombotic complex.
Protein C Control Pathway risk. 16. Describe the cause and clinical significance of
Arterial Thrombosis
8. Offer a sequence of lupus anticoagulant antibody heparin-induced thrombocytopenia (HIT).
Predictors
tests that provides the greatest diagnostic validity 17. Describe the clinical diagnosis, laboratory diagnosis,
C-Reactive Protein
Plasma Homocysteine and interpret the results of the tests. and management of HIT.
Fibrinogen Activity
Lipoprotein (a)
Disseminated
Intravascular Coagulation
Causes CASE STUDY
Pathophysiology After studying the material in this chapter, the reader should be able to respond to the following
Symptoms
case study:
Laboratory Diagnosis
Specialized Laboratory Tests A 42-year-old woman with no significant medical history developed sudden onset of shortness of
That May Aid in Diagnosis breath and chest pain. She was taken to an emergency department, where a pulmonary embolism
Treatment
was diagnosed. After admission, she was treated with intravenous heparin and given a hypercoagu-
Localized Thrombosis
lability workup.
Monitors
Heparin-Induced 1. For what conditions can the woman be tested while she is in the hospital?
Thrombocytopenia 2. List possible acquired risk factors for thrombosis that need to be excluded in this patient.
Cause and Clinical 3. What would be the implications of diagnosing a congenital risk factor for thrombosis in this
Significance patient?
Platelet Count
Other Laboratory Tests
Treatment
Conclusion *The author acknowledges the substantial contributions of Marisa B. Marques, MD, to chapter 42 of the third
edition of this textbook, many of which continue in this edition.

668
CHAPTER 42 Thrombosis Risk Testing 669
Prevalence of Venous Thrombosis
DEVELOPMENTS IN THROMBOSIS
The annual incidence of venous thrombosis in the unselected
RISK TESTING
U.S. population is 1 in 1000.7,8 This includes the comparatively
Before 1992, medical laboratory scientists performed assays to innocent thrombosis of superficial leg veins as well as the more
detect only three proven venous thrombosis risk factors: defi- dangerous deep vein thromboses that form in the iliac, popliteal,
ciencies of the coagulation control factors antithrombin, protein and femoral veins of the upper legs and calves.9 Large occlusive
C, and protein S.1 Taken together, these three deficiencies thrombi also may form, although less often, in the veins of the
accounted for no more than 7% of cases of recurrent venous upper extremities, liver, spleen, intestines, brain, and kidneys.
thromboembolic disease and bore no relationship to arterial The symptoms of thrombosis include the sensation of heat,
thrombosis. Since the report by Dahlback et al2 of activated localized pain, redness, and swelling. In deep vein thrombosis,
protein C (APC) resistance in 1993 and the characterization by the entire leg swells.
Bertina et al3 of the factor V Leiden (FVL) mutation as its cause Fragments of thrombi, called emboli, may separate from the
in 1994, efforts devoted to thrombosis prediction and evalua- proximal end of a venous thrombus, move swiftly through the
tion have redefined the hemostasis laboratory and increase its right chambers of the heart, and lodge in the arterial pulmonary
workload exponentially. The list of current assays includes APC vasculature, causing ischemia and death of lung tissue.10 Nearly
resistance, FVL mutation, prothrombin G20210A mutation, factor 95% of these pulmonary emboli arise from thrombi in the deep
VIII activity, lupus anticoagulant (LA), anticardiolipin (ACL) leg and calf veins. Of the 250,000 individuals who experience
antibodies, and anti2-glycoprotein I (anti2-GPI) for venous pulmonary embolism in the United States annually, 10% to 15%
thrombosis and tissue plasminogen activator (TPA), plasminogen die within 3 months. Many pulmonary emboli go undiagnosed
activator inhibitor 1 (PAI-1), high-sensitivity C-reactive protein because of the ambiguity of the symptoms. Coagulation system
(CRP), lipoprotein (a), plasma homocysteine, and tests for platelet imbalances, such as inappropriate activation, gain of coagula-
activity such as urinary 11-dehydrothromboxane B2 (see Chapters tion factor function, inadequate control, or fibrinolysis, are the
44 and 46) for arterial thrombosis. Technical improvements mechanisms most often implicated in venous thrombosis.11
have also been made in the diagnostic accuracy of integrated
LA identification kits, the quantitative D-dimer assay, and Prevalence of Arterial Thrombosis
tests for coagulation activation markers such as prothrombin Cardiovascular disease causes 500,000 premature deaths annu-
fragment 1+2 (PF 1+2) and thrombin-antithrombin (TAT) ally in the United States; 500,000 cerebrovascular accidents
complex.4 In addition, old tests, such as the fibrinogen assay (strokes) result in 100,000 stroke-related deaths. About 80%
and the factor VIII assay, have been applied to the prediction of myocardial infarctions and 85% of strokes are caused by
of venous and arterial thrombotic risk. thrombi that block coronary arteries or carotid end arteries of
the vertebrobasilar system, respectively.12 Transient ischemic
attacks and peripheral arterial occlusions are more frequent
INTRODUCTION TO THROMBOSIS
than strokes and coronary artery disease and, although not
Etiology of Thrombosis fatal, cause substantial morbidity.
Thrombosis is a multifaceted disorder resulting from abnormali- One important mechanism for arterial thrombosis is the
ties in blood flow, such as stasis, and abnormalities in the well-described atherosclerotic plaque formation in the vessel
coagulation system, platelet function, leukocyte activation walls. Activated platelets, monocytes, and macrophages embed
molecules, and the blood vessel wall. Thrombosis is the inap- the fatty plaque within the endothelial lining, suppressing the
propriate formation of platelet or fibrin clots that obstruct normal release of antithrombotic molecules such as nitric oxide
blood vessels. These obstructions cause ischemia (loss of blood and exposing prothrombotic substances such as tissue factor (see
supply) and necrosis (tissue death).5 Thrombophilia is defined as Chapter 40). Small unstable plaques rupture, occluding arteries
the predisposition to thrombosis secondary to a congenital or and releasing thrombotic mediators to trigger thrombotic events.
acquired disorder. The theoretical causes of thrombophilia are Activated platelets also form arterial platelet plugs with von
the following: Willebrand factor and only minimal fibrin, the white thrombi
Physical, chemical, or biologic events such as chronic or that cause death of surrounding tissue (see Chapter 13).
acute inflammation that release prothrombotic mediators The hemostasis-related lesions we associate with arterial
from damaged blood vessels or suppress blood vessel pro- thrombosis are blood vessel wall destruction and platelet acti-
duction of normal antithrombotic substances vation. Often these are inseparable. Researchers currently are
Inappropriate and uncontrolled platelet activation examining new thrombosis markers that capture pathologic
Uncontrolled triggering of the plasma coagulation system events in platelets and endothelial cells before a thrombotic
Inadequate control of coagulation-impaired fibrinolysis event occurs.

Prevalence of Thrombosis
THROMBOSIS RISK FACTORS
At least 30% of the worlds people are expected to die of
a thrombotic condition, and 25% of initial thrombotic Acquired Thrombosis Risk Factors
events are fatal.6 Many fatal thromboses go undiagnosed before In life, we acquire a legion of habits and conditions that either
autopsy. maintain or damage our hemostasis systems. Their multiplicity
670 PART VIII Hemostasis and Thrombosis

TABLE 42-1 Nondisease Risk Factors That Contribute to Thrombotic Disease


Risk Factor Comment Contribution to Thrombosis Laboratory Diagnosis
Age Thrombosis after age 50 Risk doubled each decade
Immobilization Distance driving, air travel, restriction Decreased blood flow increases thrombosis
to wheelchair or bed, obesity risk
Diet Fatty foods; inadequate folate, vitamin Homocysteinemia associated with 2-7 Plasma homocysteine, vitamin
B6, and vitamin B12 increased risk for arterial or venous levels, and lipid profile
thrombosis
Lipid metabolism imbalance Hyperlipidemia, hypercholesterolemia, Moderate thrombosis association Lipid profile: total cholesterol,
dyslipidemia, elevated lipoprotein (a), with LDL-C elevation and HDL-C, LDL-C, triglycerides,
decreased HDL-C, elevated LDL-C hypercholesterolemia, may be congenital and lipoprotein (a)
Oral contraceptive use 30mcg, formulation with progesterone 4-6 increased risk
Pregnancy 3-5 increased risk
Hormone replacement therapy 2-4 increased risk
Femoral or tibial fracture 80% incidence of thrombosis if not treated
with antithrombotic
Hip, knee, gynecologic, 50% incidence of thrombosis if not treated
prostate surgery with antithrombotic
Smoking Depends on degree hsCRP, fibrinogen
Inflammation Chronic or acute Arterial thrombosis hsCRP, fibrinogen
Central venous catheter Endothelial injury and activation 33% of children with central venous lines
develop venous thrombosis

HDL-C, High-density lipoprotein cholesterol; hsCRP, high-sensitivity C-reactive protein; LDL-C, low-density lipoprotein cholesterol.

makes it difficult to pinpoint precisely the factors that contrib- secondary to the release of procoagulant granules from the
ute to thrombosis or to determine which have the greatest malignant promyelocytes. DIC can intensify during therapy at
influence. These factors seem to contribute to venous and arte- the time of vigorous cell lysis.16 Paroxysmal nocturnal hemoglo-
rial thrombosis in varying degrees. Table 42-1 lists the nondis- binuria (see Chapter 23) is caused by a stem cell mutation that
ease risk factors implicated in thrombosis.13 modifies membrane-anchored platelet activation suppressors.
Venous or arterial thromboses occur in at least 40% of cases.17
Thrombosis Risk Factors Associated with Chronic inflammatory diseases cause thrombosis through a
Systemic Diseases variety of mechanisms, such as elevated fibrinogen and factor
In addition to life events, several conditions and diseases VIII, decreased fibrinolysis, promotion of atherosclerotic plaque
threaten us with thrombosis. Some are listed in Table 42-2, with formation, and reduced free protein S activity secondary to
an indication of the laboratorys diagnostic contribution.14 increased C4b-binding protein (C4bBP). Diabetes mellitus is a
Together, transient and chronic antiphospholipid (APL) particularly dangerous chronic inflammatory condition, raising
antibodies such as LA, ACL, and anti2-GPI may be detected the risk of cardiovascular disease sixfold. Conditions associated
in 1% to 2% of the unselected population. Chronic APL anti- with venous stasis, such as congestive heart failure, also are
bodies confer a risk of venous or arterial thrombosisa condi- common risk factors for venous thrombosis. Untreated atrial
tion called the antiphospholipid syndrome (APS). Chronic APL fibrillation increases the risk of ischemic strokes due to clot
antibodies often accompany autoimmune connective tissue formation in the right atrium and embolization to the brain.18
disorders, such as lupus erythematosus. Some appear in Nephrotic syndrome creates protein imbalances that lead to
patients without any apparent underlying disease. thrombosis through loss of plasma procoagulants. Nephrotic
Malignancies often are implicated in venous thrombosis. syndrome may also cause hemorrhage (see Chapter 41).19
One mechanism is tumor production of tissue factor analogues
that trigger chronic low-grade disseminated intravascular coag- Congenital Thrombosis Risk Factors
ulation (DIC). In addition, stasis and inflammatory effects Congenital thrombophilia is suspected when a thrombotic
increase the risk of thrombosis. Migratory thrombophlebitis, event occurs in young adults; occurs in unusual sites such as
or Trousseau syndrome, is a sign of occult adenocarcinoma such the mesenteric, renal, or axillary veins; is recurrent; or occurs in
as cancer of the pancreas or colon.15 a patient who has a family history of the disorder (Table
Myeloproliferative neoplasms such as essential thrombocythe- 42-3).20 Because thrombosis is multifactorial, however, even
mia and polycythemia vera (see Chapter 34) may trigger throm- patients with congenital thrombophilia are most likely to expe-
bosis, probably through platelet hyperactivity. A cardinal sign rience thrombotic events because of a combination of consti-
of acute promyelocytic leukemia (see Chapter 36) is DIC tutional and acquired conditions.21
CHAPTER 42 Thrombosis Risk Testing 671

TABLE 42-2 Diseases with Thrombotic Risk Components


Disease Examples or Effects Contribution to Thrombosis Laboratory Diagnosis
Antiphospholipid Chronic antiphospholipid When chronic, 1.6-3.2 increased risk of Mixing studies, lupus anticoagulant
syndrome antibody often secondary to stroke, myocardial infarction, recurrent profile, anticardiolipin antibody,
autoimmune disorders spontaneous abortion, venous thrombosis anti2-glycoprotein I immunoassays
Myeloproliferative Essential thrombocythemia, Increased risk due to plasma viscosity, Platelet counts and platelet
neoplasms polycythemia vera, chronic platelet activation lumiaggregometry
myelogenous leukemia
Hepatic and renal Diminished production or loss of Increased risk due to deranged coagulation Prothrombin time, proteins C and S, and
disorders coagulation control proteins pathways, excess thrombin production antithrombin assays, factor assays
Cancer Trousseau syndrome, low-grade 20 increased risk of thrombosis; 10-20% DIC profile: platelet count, D-dimer, Fg;
chronic DIC of people with idiopathic venous markers of coagulation activation:
thrombosis have cancer prothrombin fragment 1+2,
thrombin-antithrombin complex
Leukemia Acute promyelocytic leukemia Increased risk for chronic DIC DIC profile: platelet count, D-dimer, Fg
(M3), acute monocytic
leukemia, (M4-M5)
Paroxysmal nocturnal Platelet-related thrombosis Increased risk for deep vein thrombosis, Flow cytometry phenotyping for CD55
hemoglobinuria pulmonary embolism, DIC and CD59; DIC profile: platelet count,
D-dimer, Fg
Chronic inflammation Diabetes, infections, autoimmune Factor VIII, Fg, high-sensitivity C-reactive
disorders, smoking protein

DIC, Disseminated intravascular coagulation; Fg, fibrinogen.

TABLE 42-3 Predisposing Congenital Factors and Thrombosis Risk


Risk Factor Comment Risk of Thrombosis Laboratory Tests
AT deficiency AT inhibits the serine proteases IIa Heterozygous: increased 10-20 Clot-based and chromogenic AT
(previously called (thrombin), IXa, Xa, and XIa. Antithrombin Homozygous: 100%, rarely reported activity assays, immunoassays
AT III deficiency) function is enhanced by heparin. for AT concentration (antigen)
PC deficiency Activated PC is a serine protease that Heterozygous: increased 2-5 Clot-based and chromogenic PC
hydrolyzes factors Va and VIIIa, requires Homozygous: 100%; causes activity assays, immunoassay for
protein S as a cofactor. neonatal purpura fulminans PC concentration (antigen)
Free PS deficiency PS is a cofactor for activated protein C, 40% Heterozygous: increased 1.6-11.5 Clot-based free PS activity assays,
is free, 60% circulates bound to C4bBP. Homozygous: 100% but rarely free and total PS immunoassays
reported; causes neonatal for PS concentration (antigen)
purpura fulminans
APC resistance Factor V Leiden (T506Q) mutation gain of Heterozygous: increased 3 PTT-based APC resistance test and
function renders factor V resistant to APC. Homozygous: increased 18 confirmatory molecular assay
Prothrombin G20210A Mutation in genes untranslated 3 promoter Heterozygous: increased 1.6-11.5 Molecular assay only; phenotypic
region creates moderate elevation in assay provides no specificity
prothrombin activity.
Dysfibrinogenemia and Association is seen with arterial thrombosis. Under investigation: acute phase Fibrinogen clotting assay, thrombin
afibrinogenemia reactant time, reptilase time
Plasminogen mutations Rare cases have been described. Chromogenic substrate
Tissue plasminogen Under investigation Chromogenic substrate assay
activator deficiency
PAI-1 elevation PAI-1 increase may be common. Chromogenic substrate assay

APC, Activated protein C; AT, antithrombin; C4bBP, C4b-binding protein; PAI-1, plasminogen activator inhibitor 1; PC, protein C; PS, protein S; PTT, partial thromboplastin time.
672 PART VIII Hemostasis and Thrombosis

TABLE 42-4 Prevalence of Congenital Thrombosis Risk Factors in the General Population and in Individuals with
Recurrent Thrombotic Disease
People with at Least
Factor Unselected Population One Thrombotic Event
Activated protein C resistance, factor V Leiden mutation 3-8% of whites, absent in Asians or Africans 20-25%
Prothrombin G20210A 2-3% of whites, absent in Asians or Africans 4-8%
Antithrombin deficiency 1 in 2000 to 1 in 5000 1-1.8%
Protein C deficiency 1 in 300 2.5-5.0%
Protein S deficiency Unknown 2.8-5.0%
Hyperhomocysteinemia associated with methylenetetrahydrofolate 11% 13.1-26.7%
reductase gene mutations
Dysfibrinogenemia Unknown 1%
Abnormal fibrinolysis Unknown 2%

The antithrombin test (previously called the antithrombin III results that determine the patients cumulative risk of throm-
or AT III test) has been available since 1972, and protein C and bosis.25 The presence or absence of laboratory-detected risk
protein S activity assays became available in the mid-1980s. factors does not affect anticoagulant treatment, however, when
The 1990s brought the APC resistance assay, the confirmatory thrombosis is in progress. Current anticoagulant therapy and
FVL mutation molecular assay, the prothrombin G20210A ongoing or recent thrombotic events affect the interpretation
mutation molecular assay, and tests for dysfibrinogenemia, of antithrombin, protein C, protein S, factor VIII, and LA
plasminogen deficiency, plasma TPA, and plasma PAI-1. testing. These assays should be performed 10 to 14 days after
APC resistance is found in 3% to 8% of whites. Resistance anticoagulant therapy is withdrawn (Table 42-5).
extends to Arabs and Hispanics, but the mutation is absent
from African and East Asian populations (Table 42-4).22 APC Antiphospholipid Antibodies
resistance may exist in the absence of the FVL mutation and APL antibodies are a family of immunoglobulins that bind
occasionally is acquired in pregnancy or in association with protein-phospholipid complexes.26 APL antibodies include
oral contraceptive therapy. LAs, detected by clot-based profiles, and ACL antibodies and
The prothrombin G20210A gene mutation is the second anti2-GPI antibodies, detected by immunoassay. Chronic
most common inherited thrombophilic tendency in patients autoimmune APL antibodies are associated with APS, which is
with a personal and family history of deep vein thrombosis.23 characterized by transient ischemic attacks, strokes, coronary
All together, protein C, protein S, and antithrombin deficien- and peripheral artery disease, venous thromboembolism, and
cies are found in 0.2% to 1.0% of the world population. The repeated pregnancy complications.27,28
incidences of dysfibrinogenemia and the various forms of APL antibodies arise as immunoglobulin M (IgM), IgG, or
abnormal fibrinolysis (plasminogen deficiency, TPA deficiency, IgA isotypes. Because they may bind a variety of protein-
and PAI-1 excess) are under investigation. phospholipid complexes, they are called nonspecific inhibitors.
Their name reflects the fact that they previously were thought
Double Hit to bind phospholipids directly; however, their target antigens
Thrombosis often is associated with a combination of genetic are the proteins assembled on anionic phospholipid surfaces.29
defect, disease, and lifestyle influences. The fact that an indi- The plasma protein most often bound by APL antibodies is
vidual possesses protein C, protein S, or antithrombin defi- 2-GPI, although annexin V and prothrombin are sometimes
ciency does not mean that thrombosis is inevitable. Many implicated. APL antibodies probably develop in response to
heterozygotes experience no thrombotic event during their life- newly formed protein-phospholipid complexes, and investiga-
times, whereas others experience clotting only when two or tors still are learning exactly how they cause thrombosis.30,31
more risk factors converge. A young woman heterozygous for
the FVL mutation has a 35-fold increase in thrombosis risk upon Clinical Consequences of
starting oral contraceptive therapy. In the Physicians Health Antiphospholipid Antibodies
Study, homocysteinemia tripled the risk of idiopathic venous Between 1% and 2% of unselected individuals of both sexes and
thrombosis, and the FVL mutation doubled it. When both were all races, and 5% to 15% of individuals with recurrent venous
present, the risk of venous thrombosis was increased 10-fold.24 or arterial thrombotic disease have APL antibodies.32 Most APL
antibodies arise in response to a bacterial, viral, fungal, or para-
sitic infection or to treatment with numerous drugs (Box 42-1)
LABORATORY EVALUATION
and disappear within 12 weeks. These are mostly transient
OF THROMBOPHILIA
alloimmune APL antibodies and have no clinical consequences.33
When thrombophilia is suspected, it is important to assess all Nevertheless, the laboratory scientist must follow up any positive
known risk factors, because it is the combination of positive results on APL antibody assays to determine their persistence.
CHAPTER 42 Thrombosis Risk Testing 673

TABLE 42-5 Complete Thrombophilia Laboratory Test Profile


Assay Reference Value/Interval Comments
APC resistance Ratio 1.8 Clot-based screen based on PTT and factor Vdepleted plasma.
Factor V Leiden mutation Wild-type Molecular assay performed as follow-up to APC resistance ratio of <1.8
Prothrombin G20210A Wild-type Molecular assay. There is no phenotypic assay for prothrombin G20210A.
LA profile* Negative for LA Minimum of two clot-based assays such as PTT, DRVVT, KCT, or dilute PT
with phospholipid neutralization test for immunoglobulins of APL family.
ACL antibody IgG: <12GPL Immunoassay for immunoglobulins of APL family. ACL depends on 2-GPI
IgM: <10MPL in reaction mix.
Anti2-GPI antibody <20G units Immunoassay for an immunoglobulin of APL family. 2-GPI is key
phospholipid-binding protein in family.
AT activity* 78-126% Serine protease inhibitor suppresses IIa (thrombin), IXa, Xa, XIa. When
consistently below reference limit, follow up with AT antigen assay.
PC activity* 70-140% Digests VIIIa and Va. When consistently below reference limit, follow up
with PC antigen assay.
PS activity* 65-140% PC cofactor. When consistently below reference limit, follow up with total
and free PS antigen assay, C4b-binding protein assay.
Factor VIII activity 50-186% Confirm elevated factor VIII after 12wk.
Fibrinogen 220-498mg/dL Clot-based assay. Elevation may be associated with arterial thrombosis.
Plasminogen activator inhibitor 1 <30units/mL Elevation may be associated with venous thrombosis.

ACL, Anticardiolipin; APC, activated protein C; APL, antiphospholipid; AT, antithrombin; 2-GPI, 2-glycoprotein I; DRVVT, dilute Russell viper venom time; GPL, IgG antiphospholipid
antibody unit; Ig, immunoglobulin; KCT, kaolin clotting time; LA, lupus anticoagulant; MPL, IgM antiphospholipid antibody unit; PC, protein C, PS, protein S; PT, prothrombin time;
PTT, partial thromboplastin time.
*Inaccurate during active thrombosis or anticoagulant therapy. Perform 14 days after anticoagulant therapy is discontinued.

clinical hemostasis laboratories offer APL detection systems


BOX 42-1 Agents Known to Induce Antiphospholipid that include clot-based assays for LA and immunoassays for
Antibodies ACL and anti2-GPI antibodies. Occasionally, an LA is sus-
Various antibiotics
pected because of an unexplained prolonged partial throm-
Phenothiazine
boplastin time (PTT) that does not correct when the test is
Hydralazine
repeated in mixing studies (see Chapter 45).35,36
Quinine and quinidine
Calcium channel blockers
Lupus Anticoagulant Test Profile
Procainamide
Test systems that employ low-reagent phospholipid concentra-
Phenytoin
tions are sensitive to LA.37 There are four such systems, at least
Cocaine
two of which are required to make up an LA profile. The need
Elevated estrogens
for multiple test systems arises from the multiplicity of LA
reaction characteristics, and a confirmed positive result in one
system is conclusive despite a negative result in another. The
Autoimmune APL antibodies are part of the family of auto- four available systems are PTTs formulated with low reagent
antibodies that arise in collagen vascular diseases; 50% of phospholipid concentrations and silica-based particulate acti-
patients with systemic lupus erythematosus have autoimmune vators designed to be LA sensitive: the dilute Russell viper
APL antibodies. Autoimmune APL antibodies are also detected venom time (DRVVT), the kaolin clotting time (KCT), and the
in patients with rheumatoid arthritis and Sjgren syndrome, dilute thromboplastin time (DTT).38 As illustrated in Figure
but may arise spontaneously, a disorder called primary APS. 42-1, the KCT and PTT trigger coagulation at the level of factor
Autoimmune APL antibodies may persist, and fully 30% are XII; DRVVT, at factor X; and DTT, at factor VII (see Chapter
associated with arterial and venous thrombosis. Chronic pres- 45). The 2009 International Society on Thrombosis and Hae-
ence of an autoimmune APL antibody not associated with a mostasis (ISTH) update of guidelines for LA detection recom-
known underlying autoimmune disorder confers a 1.8-fold to mends using the DRVVT and LA-sensitive PPT as the two
3.2-fold increased risk of thrombosis. primary screening tests (Figures 42-2 and 42-3). This combina-
tion of assays updates the following 1995 ISTH requirements
Detection and Confirmation of for LA identification39:
Antiphospholipid Antibodies 1. Prolonged phospholipid-dependent clot formation using a
Clinicians suspect APS in unexplained venous or arterial throm- screen such as the low phospholipid PTT, DRVVT, KCT,
bosis, thrombocytopenia, or recurrent fetal loss.34 Specialized or DTT
674 PART VIII Hemostasis and Thrombosis

2. Failure to correct the prolonged clot formation by mixing phospholipids. Platelet membrane fragments formed during
with normal platelet-poor plasma and repeating the test freezing and thawing can likewise neutralize LA and lead to a
(mixing study) false-negative LA result.
3. Shortening or correction of the prolonged screen test result
by addition of excess phospholipids Performing the Clot-Based Lupus Anticoagulant
4. Exclusion of other coagulopathies Mixing Study. When the PTT or DRVVT is prolonged and
In performing mixing studies, it is essential to use only anticoagulant therapy, particularly treatment with unfraction-
platelet-poor plasma (plasma centrifuged so that it has a plate- ated heparin, has been ruled out, a new aliquot of patient
let count of less than 10,000/mcL). The use of platelet-poor platelet-poor plasma is mixed 1:1 with pooled normal platelet-
plasma avoids neutralization of LA by the platelet membrane poor plasma, and the assay is repeated on the mixture using
the same test system (immediate mix). Some LAs are time and
temperature dependent, so if the result for the mixture shows
KCT and PTT correction to within 10% or to within 5 seconds of the result
for the normal platelet-poor plasma, the test should be repeated
DPT Vlla Xla Xlla
again after incubating the mix at 37 C for 1 to 2 hours. Each
TF Pre-K HMWK laboratory manager or director must decide what degree of
shortening constitutes correction. Many use the Rosner index,
IXa Vllla which defines correction as a mixture result within 10% of
the result of the platelet-poor normal plasma.40 Many others
define correction as return to a value within the PTT reference
DRVVT Xa Va interval.
If either the PTT or the DRVVT remains prolonged (that is,
uncorrected), the presence of LA is presumed, and confirma-
Thr Xllla tion is necessary. Confirmatory tests employ the same assay
that originally yielded prolonged results, adding a high-
Cross-linked concentration phospholipid reagent such as hexagonal phase
Fibrinogen Fibrin polymer
fibrin phospholipid. If LA was the cause of prolongation, the high-
Figure 42-1 Kaolin clotting time (KCT), partial thromboplastin time (PTT), phospholipid test result shortens (corrects).41
dilute prothrombin time (DPT), and dilute Russell viper venom time (DRVVT).
The simplified coagulation mechanism is used to illustrate that the KCT and Anticardiolipin Antibody Immunoassay
PTT initiate coagulation at the level of factor XII, DRVVT at the level of factor LA and ACL antibodies coexist in 60% of cases, and both may
X, and DPT at the level of factor VII. be found in APS.42 The ACL test is an immunoassay that may

PTT long Mix test plasma PTT mix


1:1 with NP, incubate, No correction
PTT on mixture
Thrombin time TT norm

Phospholipid
TT long1 PTT mix Neutralization
corrects
PTT
Hepzyme long Positive2 Negative

No LA
Repeat PTT Suspect factor
deficiency LA No LA

PTT
norm 1. If the DRVVT is normal, proceed to the Hepzyme step. If the DRVVT is
prolonged, Hepzyme is unnecessary.
2. If phospholipid neutralization is positive and DRVVT negative, assay FVIII
No LA to determine if the positive neutralization step is due to a FVIII inhibitor.

Figure 42-2 Lupus anticoagulant (LA) detection using an LA-sensitive partial thromboplastin time (PTT) system. Use in conjunction with the dilute Russell
viper venom time (DRVVT) detection system (see Figure 42-3). If the LA-sensitive PTT is prolonged but the DRVVT is normal (norm), measure thrombin time
(TT) to detect heparin. If the TT is prolonged, treat the plasma with Hepzyme and repeat the PTT test. If the PTT is still prolonged, or if no heparin was present,
mix the patient plasma with pooled platelet-free normal plasma (NP), incubate 1 to 2 hours, and repeat. If the PTT mix result is corrected, suspect a factor
deficiency and confirm. If the PTT for the mix is prolonged, proceed to the phospholipid neutralization step to confirm LA. If the phospholipid neutralization
result is positive but the DRVVT result is negative, perform a factor VIII assay to determine if the findings of the phospholipid neutralization step are due to a
factor VIII inhibitor.
CHAPTER 42 Thrombosis Risk Testing 675

Mix test plasma


1:1 with NP, incubate, DRVVT mix
DRVVT long
DRVVT on mixture No correction

DRVVT confirmation Negative

DRVVT mix
corrects
Positive
No LA

LA
No LA
Suspect factor
deficiency

Figure 42-3 Lupus anticoagulant (LA) detection using the dilute Russell viper venom time (DRVVT) system. Use in conjunction with the LA-sensitive partial
thromboplastin time detection system (see Figure 42-2). If the DRVVT is prolonged, mix the patient plasma with pooled platelet-free normal plasma (NP),
incubate 1 to 2 hours, and repeat. If the DRVVT for the mix is normal, suspect a factor deficiency and confirm. If the DRVVT for the mix is prolonged, proceed
to the phospholipid neutralization step to confirm.

be normalized among laboratories and is not affected by


Negative Negative
heparin therapy, oral anticoagulant therapy, current thrombo- ACL
sis, or factor deficiencies. Anti-2GPI APTS No APL
The manufacturer coats microplate wells with bovine heart
cardiolipin and blocks (fills open receptor sites) with a bovine Negative
Positive
serum solution containing 2-GPI. Test sera or plasmas are
serially diluted and pipetted to the wells along with calibrators
APL transient
and controls. ACL binds the solid-phase cardiolipin2-GPI Repeat positive assay after 12 weeks not significant
target complex and cannot be washed from the wells. Enzyme-
labeled antihuman IgG, IgM, or IgA conjugates are added. A
color-producing substrate is then added, and a color change
indicates the presence of ACL. Color intensities of the patient APL chronic, clinically significant
and control samples are compared with the calibrator curve. Figure 42-4 Reflexive immunoassay sequence for antiphospholipid (APL)
Results are expressed using GPL, MPL, or APL units, where 1 antibodies. If either the immunoglobulin M (IgM) or IgG anticardiolipin (ACL)
unit is equivalent to 1mcg/mL of an affinity-purified standard immunoassay or the IgM or IgG anti2-glycoprotein I (anti2-GPI) immu-
IgG, IgM, or IgA specimen.43 Reference limits are established noassay yields a positive result, confirm to establish chronicity after 12
in each laboratory. weeks. If either is confirmed, the patient has a clinically significant APL
antibody. If the initial ACL and anti2-GPI results are negative but an APL
Anti2-Glycoprotein I Immunoassay antibody is suspected, perform the antiphosphatidylserine (APTS) immunoas-
IgM and IgG anti2-GPI immunoassays are performed as a say. If the result is positive, repeat after 12 weeks to establish chronicity.
part of the profile that includes ACL assays.44 An anti2-GPI
result of greater than 20GPL or MPL units correlates with
thrombosis more closely than the presence of ACL antibodies. Activated Protein C Resistance and
Any ACL or 2-GPI assay yielding positive results should be Factor V Leiden Mutation
repeated on a new specimen collected after 12 weeks to distin- Clinical Importance of
guish a transient alloantibody from a chronic autoantibody Activated Protein C Resistance
(Figure 42-4). Activated factors V and VIII (factors Va and VIIIa) are inacti-
vated by the APCprotein S complex. A mutation in the factor
Antiphosphatidylserine Immunoassay V gene substitutes glutamine for arginine at position 506 of the
For cases in which an APL antibody is suspected but the factor V molecule (FV R506Q). The arginine is a cleavage site
routine LA, ACL, and 2-GPI assay results are negative, the for APC, so the substitution slows or prevents hydrolysis of the
clinician may wish to order the antiphosphatidylserine immu- factor V molecule (Figure 42-5). Resistant factor Va remains
noassay to detect APL antibodies specific for phosphatidyl active and increases the production of thrombin. The factor V
serine.45 A result greater than or equal to 16IgG or 22IgM R506Q mutation is named for the city in the Netherlands in
antiphosphatidylserine units is considered positive. The anti which it was first described, Leiden (FVL mutation). Between
phosphatidylserine assay is available from specialty reference 3% and 8% of Northern European whites possess FVL (see
laboratories. Table 42-4).46 Owing to its prevalence and the associated 3-fold
676 PART VIII Hemostasis and Thrombosis

Performance Characteristics of the Activated Protein


A1 A2 B A3 C1 C2
C Resistance Test. Factor Vdepleted plasma compensates
for potential factor deficiencies and for oral anticoagulant
APC cleavage site
therapy by providing procoagulant cofactors. The laboratory
scientist uses only platelet-poor plasma in the APC resistance
Normal 504 506 508 test to prevent loss of sensitivity caused by the abundant release
of platelet factor V. Most APC resistance reagent kits contain
L D R R G I Q polybrene or heparinase to neutralize heparin. LA affects the
test system adversely, however.50 If LA is present, the molecular
R506Q 504 506 508 test for FVL is indicated.51
L D R Q G I Q
Factor V Leiden Mutation Assay
Most laboratories confirm the APC resistance diagnosis using
No cleavage
the molecular FVL mutation test. The determination of zygosity
Figure 42-5 Factor V Leiden mutation. A point mutation of the factor V is important to predict the risk for thrombosis.
gene results in the substitution of glutamine for arginine at position 506
(R506Q) of the factor V molecule. The normal arginine at position 506 is a
Prothrombin G20210A
cleavage site for activated protein C (APC), so the substitution slows or
A guanine-to-adenine mutation at base 20210 of the 3 untrans-
prevents cleavage of the factor V molecule. D, Aspartic acid; G, glycine; I,
isoleucine; L, leucine; R, arginine, Q, glutamine. lated region of the prothrombin (factor II) gene has been asso-
ciated with mildly elevated plasma prothrombin levels,
averaging 130%.52 The increased risk of thrombosis in those
with the mutation seems to be related to the elevated pro-
PTT reagent, factor Vdepleted plasma thrombin activity.53 The prevalence of this mutation among
+ patient plasma; two aliquots of mixture
individuals with familial thrombosis is 5% to 18%, whereas
prevalence worldwide is 0.3% to 2.4%, depending on race.54
The risk of venous thrombosis in heterozygotes is only two to
Add CaCl2 Add CaCl2 + APC three times the baseline risk. Although the mutation may cause
a slight elevation in test results, a phenotypic prothrombin
activity assay is of little diagnostic value because there is con-
siderable overlap between normal prothrombin levels and pro-
Result: 27 seconds Normal Result: >49 seconds
thrombin levels in people with the mutation.

Result: 27 seconds APCR Result: <49 seconds Antithrombin


Figure 42-6 Measurement of activated protein C resistance ratio (APCR) Antithrombin is a serine protease inhibitor (serpin) that neu-
using factor Vdepleted plasma. Patient plasma is mixed 1:4 with factor tralizes factors IIa (thrombin), IXa, Xa, XIa, and XIIa. Anti-
Vdepleted plasma and partial thromboplastin time (PTT) reagent. Two ali- thrombin activity is enhanced by unfractionated heparin,
quots of this mixture are then tested: one aliquot is combined with calcium low-molecular-weight heparin, and synthetic pentasaccharide
chloride (CaCl2) alone, and the other aliquot is mixed with CaCl2 plus activated (Figures 42-7 and 42-8). Antithrombin was the first of the
protein C (APC). The mixture with the APC should be slowed to give a clotting plasma coagulation control proteins to be identified and the
time at least 1.8 times longer than the mixture without APC. A ratio of less first to be assayed routinely in the clinical hemostasis labora-
than 1.8 implies APC resistance. tory.55 Other members of the serpin family are heparin cofactor
II, 2-macroglobulin, 2-antiplasmin, and 1-antitrypsin. Typi-
cally, hemostasis specialty laboratories or reference laborato-
higher thrombosis risk (18-fold higher for homozygotes), the ries are the only places that offer serpin assays other than the
acute care hemostasis laboratory must provide APC resistance more common antithrombin activity assay, which has found
detection to screen for FVL.47 its way into laboratories at most acute care facilities.

Activated Protein C Resistance Clot-Based Assay Antithrombin Deficiency


In the APC resistance clot-based assay, patient plasma is mixed Acquired antithrombin deficiency occurs in liver disease, in
1:4 with factor Vdepleted plasma.48 PTT reagent is added to nephrotic syndrome, with prolonged heparin therapy, with
two aliquots of the mixture and incubated for 3 minutes asparaginase therapy, with the use of oral contraceptives, and
(Figure 42-6). A solution of calcium chloride is pipetted into in DIC, where antithrombin is rapidly consumed. Congenital
one mixture, and the clot formation is timed. A solution of deficiency is present in 1 in 2000 to 1 in 5000 of the general
calcium chloride with APC is added to the second mixture and population and accounts for 1.0% to 1.8% of recurrent venous
clotting is timed. The normal ratio of PTT results between the thromboembolic disease.56 About 90% of cases of antithrom-
two assays is 1.8 or greater. In APC resistance, the ratio is less bin deficiency are quantitative (reduced production), or type
than 1.8.49 1; the remainder are caused by mutations creating structural
CHAPTER 42 Thrombosis Risk Testing 677

Protease Active Reagent


binding protease heparin
site site
AT AT
AT Ila 

Heparin Heparin Measured reagent Xa


binding site binding site
AT Xa
Heparin with AT Ila AT  Xa
17 sugar units

Excess Xa
AT Ila
AT Xa
Figure 42-7 The reaction between antithrombin (AT) and activated coagu-  Xa
lation factor II (IIa), or thrombin, is supported by unfractionated heparin.
Standard unfractionated heparin, with a molecular weight of 5000D to
40,000D, provides polysaccharide chains of at least 17 sugar subunits. Substrate Product
Heparin molecules of this length support the reaction between AT and IIa, as Figure 42-9 Chromogenic antithrombin (AT) functional assay. Patient
the antithrombin and IIa assemble on the heparin molecule. The AT molecule plasma is pipetted into a reagent consisting of heparin, a measured concen-
attaches to a specific pentasaccharide sequence. The IIa possesses a heparin tration of activated coagulation factor X (Xa), and a chromogenic substrate.
binding site that enables it to assemble on the heparin surface adjacent to AT is activated by heparin and binds Xa. Excess Xa hydrolyzes the substrate
the AT, a property called approximation. The AT is sterically modified (allo- and produces a yellow product, para-nitroaniline (pNA). The intensity of pNA
stery), supporting a covalent reaction between the AT protease binding site color is inversely proportional to AT activity.
and the IIa active protease site. Thrombin (IIa) and antithrombin, covalently
bound, release from heparin and form measurable plasma thrombin-
antithrombin (TAT) complexes, useful as a marker of coagulation activation. Antithrombin antigen levels range from 22 to 39mg/dL (68%
to 128%) by latex microparticle immunoassay, and the plasma
biologic half-life is 72 hours. Adult levels are reached by 3
Protease Active
binding protease months of age, and levels remain steady throughout adult life
site site except during periods of physiologic challenge, such as preg-
nancy. Antithrombin activity decreases with age.
AT Xa

Antithrombin Activity Assay


Heparin No heparin
binding site binding site
Screening for antithrombin deficiency should be performed
using a clot-based or chromogenic functional assay. Many
Heparin with AT Xa laboratories choose the chromogenic assay because of its sta-
~7 sugar units
bility and reproducibility. Test plasma is mixed with heparin
and a protease, usually factor Xa (Figure 42-9). The mixture is
incubated at 37 C for several minutes. During incubation, the
AT Xa heparin-activated antithrombin irreversibly binds a proportion
Figure 42-8 The reaction between antithrombin (AT) and activated coag- of the reagent protease. Chromogenic substrate, provided as a
ulation factor X (Xa) is supported by low-molecular-weight heparin. Low- second reagent, is hydrolyzed by residual protease. The degree
molecular-weight heparin, with a molecular weight of less than 8000D, of hydrolysis, measurable by colored end product intensity, is
provides polysaccharide chains of six or seven sugar subunits. Heparin inversely proportional to the activity of antithrombin in the
molecules of this length support the reaction between AT and Xa. The AT test plasma.57 The chromogenic substrate test for plasma anti-
molecule attaches to a specific pentasaccharide sequence. The Xa does not thrombin activity detects quantitative and qualitative anti-
possess a heparin binding site and does not need to assemble on the heparin
thrombin deficiencies and detects mutations affecting the
surface adjacent to the AT. The AT is sterically modified (allostery), supporting
proteolytic site, but not the heparin binding site. Clot-based
a covalent reaction between the AT protease binding site and the Xa active
protease site. antithrombin assays, although less popular, are affected by,
and therefore detect, mutations in both the proteolytic and
heparin binding sites, but may not provide the precision of the
abnormalities in the protease binding site or the heparin binding chromogenic substrate assay.
site. Type 2 mutations do not reduce antithrombin production,
although the molecules are nonfunctional. Antithrombin Antigen Assay
Antithrombin concentration is measured in a turbidometric
Antithrombin Reference Ranges microparticle immunoassay using a suspension of latex micro-
Adult plasma antithrombin activity ranges from 78% to beads coated with antibody. In the absence of antithrombin,
126%, whether measured by clot-based or chromogenic assay. the wavelength of incident monochromatic light exceeds the
678 PART VIII Hemostasis and Thrombosis

latex microparticle diameter, so the light passes through unab- Endothelial


sorbed. In the presence of antithrombin, the particles form cell membrane
larger aggregates. The antithrombin concentration is directly
proportional to the rate of light absorption change.58 Anti- TM Thrombin
thrombin antigen levels are diminished in quantitative (type
1) but not qualitative (type 2) antithrombin deficiency. Oral Endothelial cell or
Va VIIIa
anticoagulant warfarin therapy may raise the antithrombin platelet membrane
level and mask mild deficiency. Antithrombin activity remains
decreased for 10 to 14 days after surgery or a thrombotic event, PC APC PS
so the assay should not be used to establish a congenital defi-
ciency during this period.

Free PS 40% Vi VIIIi


Heparin Resistance and the Antithrombin Assay
Antithrombin may be decreased during prolonged or intense
heparin therapy and may be totally consumed if the patient
has a congenital antithrombin deficiency. In this instance,
PS Bound PS 60%
heparin may be administered in therapeutic or higher dosages,
but it neither exerts an anticoagulant effect nor is detected by
C4bBP
the PTT. This is known as heparin resistance. In such cases, an
antithrombin assay is necessary to confirm antithrombin defi- Figure 42-10 Protein C pathway. Thrombin (activated coagulation factor
ciency. Antithrombin deficiency may be treated with anti- II, or IIa) binds constitutive thrombomodulin (TM) on the endothelial cell
thrombin concentrate (Thrombate III; Alecris Biotherapeutics, membrane. The TM-thrombin complex activates protein C (PC). Activated
Inc., Research Triangle Park, N.C.).59 protein C (APC) binds protein S (PS) on endothelial cell or platelet membrane
phospholipids. The bound active complex hydrolyzes activated coagulation
Protein C Control Pathway factors V and VIII (Va and VIIIa). C4bBP, C4b-binding protein; Vi, inactivated
Thrombin is an important coagulation factor because it cleaves coagulation factor V; VIIIi, inactivated coagulation factor VIII.
fibrinogen and activates platelets and factors V, VIII, XI, and
XIII. In the intact vessel where clotting would be inexpedient,
thrombin binds endothelial cell membrane thrombomodulin half-life of protein C is 6 hours, its level decreases as rapidly as
and becomes an anticoagulant.60 How does this paradox that of factor VII at the start of warfarin therapy. In heterozy-
happen? The thrombin-thrombomodulin complex activates gous protein C deficiency, protein C activity may decrease to a
plasma protein C, and APC binds free plasma protein S (Figure thrombotic (procoagulant) level (less than 65%) more rapidly
42-10). The stabilized APCprotein S complex hydrolyzes than the coagulation factors reach a safe anticoagulant level
factors Va and VIIIa to slow coagulation. Recurrent venous (less than 30%). Consequently, in early warfarin therapy a
thrombosis is the potential consequence of any deficiency in patient may experience warfarin-induced skin necrosis, a para-
this complex pathway. doxical situation in which the anticoagulant therapy brings on
Protein S, the cofactor that binds and stabilizes APC, circu- thrombosis of the dermal vessels. This complication is sus-
lates in the plasma in two forms: free and covalently bound to pected when the patient develops painful necrotic lesions that
the complement control protein C4bBP. Bound protein S are preceded by itching. The necrosis may require surgical
cannot participate in the protein C anticoagulant pathway; dbridement. To avoid this risk, administration of low-
only free plasma protein S is available to serve as the APC molecular-weight heparin or synthetic pentasaccharide is
cofactor. Protein SC4bBP binding is of particular interest in recommended along with warfarin until a satisfactory and
inflammatory conditions, where the plasma C4bBP level rises, stable international normalized ratio (INR) is reached (see
binding additional protein S. Free protein S levels are propor- Chapter 46).
tionally decreased. Activity levels of protein C and protein S remain below
normal for 10 to 14 days after cessation of warfarin therapy.
Protein C and Protein S Reference Ranges Similarly, for several days after surgery or a thrombotic event,
Heterozygous deficiency of protein C or protein S leads to a these proteins are diminished even if warfarin has not been
1.6-fold to 11.5-fold increased risk of recurrent deep vein used.62 Their activities are depressed in pregnancy, liver or renal
thrombosis and pulmonary embolism.61 Protein S deficiency disease, vitamin K deficiency, and DIC. Protein C and protein
also has been implicated in transient ischemic attacks and S assays therefore cannot be used to identify a congenital defi-
strokes, particularly in the young. The reference interval for ciency when they are employed soon after thrombosis or soon
both protein C and protein S activity and antigen levels is 65% after the cessation of warfarin therapy, during pregnancy, or in
to 140%, and levels ordinarily remain between 30% and 65% the presence of DIC, liver disease, renal disease, or vitamin K
for heterozygotes. deficiency.63
Control proteins C and S (and Z) and factors II (prothrom- Homozygous protein C or protein S deficiency results in
bin) VII, IX, and X are vitamin K dependent. Because the neonatal purpura fulminans, a condition that is rapidly fatal
CHAPTER 42 Thrombosis Risk Testing 679

S-2366 RVV reagent

Dilute APC
plasma PC APC Patient plasma diluted 1:10,
unknown PS activity

Agkistrodon pNA
PS-depleted plasma
venom (yellow)
activator Clot

Figure 42-11 Chromogenic protein C (PC) assay. Patient plasma is pipet- Figure 42-12 Clot-based protein S (PS) assay. Patient plasma is diluted
ted into a reagent composed of Agkistrodon contortrix venom activator and and pipetted into a reagent composed of PS-depleted plasma. A second
chromogenic substrate (S-2366) specific to activated protein C (APC). The reagent is composed of Russell viper venom (RVV), which activates clotting
APC hydrolyzes the substrate to produce para-nitroaniline (pNA), a yellow at the level of factor X, and activated protein C (APC). The patient PS binds
product. The intensity of color is proportional to APC activity. the reagent APC to prolong the clotting time. Clotting time is proportional to
PS activity.

when left untreated. Treatment includes administration of color development after the sequential addition of peroxidase-
factor concentrates and lifelong oral anticoagulant therapy. conjugated antihuman protein C and orthophenylenediamine
substrate. The protein C antigen concentration assay detects
Protein C Assays most acquired deficiencies and quantitative congenital defi-
Functional assays detect both quantitative and qualitative ciencies, but does not detect qualitative congenital abnormali-
protein C deficiencies.64 Chromogenic or clot-based assays are ties, which is why it is used only in response to an abnormally
available for this purpose. In the former, the patients plasma reduced protein C functional assay result.
is mixed with Protac (Centerchem, Inc., Norwalk, Conn.),
derived from the venom of the southern copperhead, Agkistro- Protein S Assays
don contortrix, which activates protein C. Subsequently, a chro- As with testing for antithrombin and protein C deficiencies,
mogenic substrate is added, and its hydrolysis by the recently protein S deficiency screening requires a functional assay. No
generated APC is measured by assessing the amount of colored chromogenic assay is available.65 A clot-based assay is per-
product. The intensity of color is proportional to the activity of formed by mixing the patients plasma with protein Sdepleted
protein C in the test plasma (Figure 42-11). The assay detects normal plasma to ensure normal levels of all other factors. APC
abnormalities that affect the molecules proteolytic properties and Russell viper venom in a buffer that contains a heparin
(active serine protease site), but misses those that affect protein neutralizer are added, followed by calcium chloride, and the
Cs phospholipid binding site or protein S binding site. In cases interval to clot formation is measured. A calibration curve is
in which protein C chromogenic assays and immunoassays produced, and the more prolonged the test result, the higher
generate normal results, but the clinical condition continues to the protein S activity (Figure 42-12). The clottable protein S
indicate possible protein C deficiency, a clot-based protein C assay can be automated.
assay may detect abnormalities at these additional sites on the Therapeutic heparin levels greater than 1 unit/mL con-
molecule. sume the heparin neutralizer and lead to overestimation of
The clot-based protein C assay is based on the ability of APC protein S activity. APC resistance, LA, and the presence of
to prolong the PTT. Test plasma is mixed with protein therapeutic direct thrombin inhibitors such as argatroban
Cdepleted normal plasma to ensure normal levels of all factors may prolong the PTT and falsely raise the activity levels on
but protein C. PTT reagent mixed with Protac and a heparin clot-based protein S assays.66 Coagulation factor VII activa-
neutralizer is added, followed by calcium chloride, and the tion may occur during prolonged refrigeration of plasma at
interval to clot formation is measured. Prolongation is propor- 4 C to 10 C; this may cause underestimation of protein S
tional to plasma protein C activity. The clot-based protein C activity.67
activity assay may be performed using an automated coagula- When there is clinical suspicion of primary protein S defi-
tion analyzer. Therapeutic heparin concentrations greater than ciency based on low activity level, enzyme immunoassays are
1IU/mL consume the heparin neutralizer, prolong the PTT, employed to measure both total and free protein S antigen.
and lead to overestimation of protein C. APC resistance, LA, These assays detect most quantitative congenital deficiencies
and the presence of therapeutic direct thrombin inhibitors such and aid in the diagnosis of qualitative type II congenital
as argatroban may prolong the PTT and falsely raise the protein deficiencies characterized by normal antigen but decreased
C activity level on clot-based assays. activity of protein S. In type III deficiency, the concentration
Enzyme immunoassay is used to measure protein C antigen of free antigen protein S and its activity, but not the total
when the functional activity is low and acquired causes have antigen, are reduced (Table 42-6). The concentration of plasma
been ruled out. Microtiter plates coated with rabbit anti C4bBP, measured with an immunoassay available for research
human protein C antibody are used to capture test plasma use only, aids in the classification of the type of protein S
protein C, and the concentration of antigen is measured by deficiency.
680 PART VIII Hemostasis and Thrombosis

TABLE 42-6 Anticipated Protein S (PS) Test Results in Qualitative and Quantitative Deficiencies
Type of PS Deficiency PS Activity PS Free Antigen PS Total Antigen C4bBP
I Quantitative <65% <65% <65% Normal
II Qualitative <65% >65% >65% Normal
III Inflammation <65% <65% >65% Elevated

C4bBP, C4b-binding protein.

TABLE 42-7 Markers of Arterial Thrombosis Risk


Marker Reference Interval Comments
High-sensitivity C-reactive protein <0.55mg/L Marker of inflammation; report in relative risk ranges; stable, reproducible
Fibrinogen 220-498mg/dL >300mg/dL increases thrombotic risk; high correlation with risk;
inadequate reproducibility with numerous test platforms
Homocysteine 4.6-12.1mol/L Reference interval and predictive values vary with population; may be
reduced through vitamin B6, B12, and folate supplementation
Total cholesterol <200mg/dL Reproducible; some relationship with diet, exercise; risk prediction is not
independent of inflammation
Ratio of total cholesterol to high-density lipoprotein <10 Reproducible; elevated ratio has some relationship with diet, exercise;
cholesterol risk prediction is not independent of inflammation
Low-density lipoprotein cholesterol <130mg/dL Reproducible; may be significantly lowered with statin therapy
Lipoprotein (a) 2.2-49.4mg/dL Varies with race and age; may be lowered with statin therapy; inadequate
reproducibility
Plasminogen, tissue plasminogen activator, Available from hemostasis reference laboratories
plasminogen activator inhibitor 1, 2-antiplasmin

ARTERIAL THROMBOSIS PREDICTORS


C-Reactive Protein
Arterial thrombotic disease in the form of peripheral There is a clear correlation between the acute phase reactant
vascular disease, myocardial infarction (heart attack), and cere- CRP and myocardial infarction and stroke.70
brovascular disease (stroke) arises from atherosclerosis. The
traditional predictors of arterial thrombosis risk are elevated High-Sensitivity C-Reactive Protein Measurement
total cholesterol and low-density lipoprotein cholesterol (LDL- Plasma CRP increases several hundredfold in response to
C), or a high ratio of total cholesterol to high-density lipopro- acute inflammatory stimuli. The increase remains stable over
tein cholesterol (TC:HDL-C) secondary to deficient HDL-C. several days in vivo, and the protein is resistant to in vitro
One third of primary cardiovascular and cerebrovascular events degradation.71 The time-honored visual flocculation assays
occur in patients whose lipid profiles are normal, however, and used for CRP measurement in acute or chronic inflammatory
half of people with proven lipid risk factors never experience illness detect only gross elevations. The monoclonal antibody
a thrombotic event.68 based high-sensitivity CRP enzyme immunoassayor its
Researchers have sought additional arterial thrombosis pre- automatable rapid counterpart, the monoclonal latex micro
dictors by analyzing lipoprotein subtypes and by performing particle assaydetects normal or slightly elevated CRP concen-
prospective randomized studies of markers of inflammation. trations. Consequently, the high-sensitivity CRP assay is an
The results of these studies have led to the identification of established clinical tool to evaluate subtle chronic systemic
numerous markers of arterial thrombosis risk (Table 42-7), as inflammation and predict cardiovascular or cerebrovascular
detailed in this section. The fibrinolytic pathway markers plas- disease.72
minogen, 2-antiplasmin, and PAI-1 also are useful. Other Data from at least 10 studies involving tens of thousands of
proteins, such as TPA, show marked diurnal variation and individuals show that mild chronic elevations of CRP may
require a specialized phlebotomy protocol using a citrate, predict atherosclerosis 6 or more years before myocardial
theophylline, adenosine, dipyridamole (CTAD) tube to prevent in infarction or stroke.73 The high-sensitivity CRP procedure
vitro elevation immediately after venipuncture.69 Homocyste- enhances cardiac risk assessment independently of and in con-
ine, fibrinogen, and high-sensitivity CRP are well-established junction with measurement of lipid profile markers and is
assays with considerable predictive power. available on automated coagulation analyzers.74
CHAPTER 42 Thrombosis Risk Testing 681

TABLE 42-8 Relative Risk for Myocardial Infarction or intake and levels of vitamin B6, vitamin B12, and folate. Its
Stroke at Four Levels of High-Sensitivity C-Reactive Protein concentration is regulated by three enzymes: cystathionine
(hsCRP) Independent of Lipid Levels -synthase, which converts homocysteine to cystathionine in the
presence of vitamin B6; 5,10-methylenetetrahydrofolate reductase
Quartile hsCRP Men Women
(MTHFR), required for the remethylation of homocysteine to
1 0.55mg/L 1.0 1.0 methionine in the folic acid cycle; and methionine synthase,
2 0.56-1.14mg/L 1.8 2.9 which requires vitamin B12 (Figure 42-13). Vitamin deficiencies
3 1.15-2.10mg/L 2.5 3.5 and a functional mutation in the MTHFR gene or an inherited
4 2.11mg/L 2.9 5.5 deficiency of either cystathionine -synthase or methionine
synthase result in increased plasma homocysteine, called homo-
cysteinemia.78 Although cystathionine -synthase and methio-
nine synthase deficiencies are rare, 50% of the North American
TABLE 42-9 Relative Risk for Myocardial Infarction at
population is heterozygous for the MTHFR polymorphism.
Three Levels of High-Sensitivity C-Reactive Protein (hsCRP)
Related to Total Cholesterol
Clinical Significance of Homocysteinemia
TOTAL CHOLESTEROL Fasting homocysteinemia is an independent risk factor for arte-
Low: Medium: High: rial thrombosis, with relative risk ratios of 1.7 for coronary
hsCRP 191mg/dL 192-223mg/dL 224mg/dL artery disease, 2.5 for cerebrovascular disease, and 6.8 for
peripheral artery disease. Mild elevations of homocysteine are
Low: 0.72mg/L 1.0 1.4 2.3
an independent risk factor for occlusive arterial disease.79,80
Medium: 1.2 1.5 4.3
Homozygosity for the MTHFR G677T mutation is associated
0.73-1.69mg/L
with homocysteinemia, but is not an independent thrombosis
High: 1.70mg/L 1.1 2.3 5.3
risk factor. Several theories link homocysteinemia with coro-
nary artery disease, most citing damage to the endothelial cell.81

TABLE 42-10 Relative Risk for Myocardial Infarction at Homocysteine Test Principles
Three Levels of High-Sensitivity C-Reactive Protein (hsCRP) High-performance liquid chromatography has been a familiar
Related to Ratio of Total Cholesterol to High-Density approach to homocysteine analysis. The plasma is first treated
Lipoprotein Cholesterol (TC:HDL-C Ratio) with a reducing agent to convert all disulfide forms to homo-
cysteine, which becomes bound to fluorescein to form a
TC:HDL-C RATIO detectable fluorescent derivative. High-performance liquid
Low: Medium: High: chromatography methods are not standardized among clinical
hsCRP 3.78 3.79-5.01 5.02 laboratories.
Low: 0.72mg/L 1.0 1.2 2.8 An enzyme immunoassay is available from several dis
Medium: 0.73-1.69mg/L 1.1 2.5 3.4 tributors. In the Abbott AxSYM assay (Abbott Laboratories,
High: 1.70mg/L 1.3 2.8 4.4 Abbott Park, Ill.), plasma disulfides are reduced, the homo
cysteine is converted to S-adenosylhomocysteine, and the
S-adenosylhomocysteine is bound to an antibody conjugated
to a fluorescein tracer. This method may be adapted for auto-
High-Sensitivity C-Reactive Protein Reference mated analysis.
Range and Relative Risk When blood is collected for homocysteine measurement,
The high-sensitivity CRP mean for people who have no coro- the serum or plasma must be separated from the red blood
nary artery disease, as proven by angiography, is 0.87mg/L. In cells within minutes to avoid red blood cell release of
contrast, the mean for patients with three atherosclerotic arter- homocysteine. The serum or plasma must be kept cold and
ies is 1.43mg/L. When the high-sensitivity CRP assay is used, protected from light. Plasma homocysteine rises by 10%
CRP levels may predict the risk of stroke independent of lipid per hour if blood is kept intact at room temperature after col-
concentrations, as shown in Table 42-8. CRP levels serve as lection. Plasma collection may be accomplished using heparin,
strong predictors of risk of myocardial infarction when com- sodium citrate, or ethylenediaminetetraacetic acid (EDTA)
bined with total cholesterol (Table 42-9) or with the TC:HDL-C anticoagulants.
ratio (Table 42-10).75
Homocysteine Reference Range and Therapy
Plasma Homocysteine The reference ranges for homocysteine differ for men and
Homocysteine is a naturally occurring sulfur-containing amino women, and they rise with age, as shown in Table 42-11.
acid formed in the metabolism of dietary methionine.76,77 Therapy usually includes administration of the appropriate
Homocysteine circulates in the form of various disulfides, vitamin (folate, vitamin B6, or vitamin B12) to correct for the
together called total homocysteine. The concentration of homo- dietary deficiency or the abnormal methionine metabolism.
cysteine in plasma depends primarily on adequate protein Folate supplementation of grain in the United States, instituted
682 PART VIII Hemostasis and Thrombosis

Diet Diet

Folate Methionine
Methionine adenosyl
transferase
Adenosyl
Methionine
Tetrahydrofolate
Methyltransferase
Methylenetetrahydrofolate Methionine synthase
reductase Vitamin B12
Adenosyl
5-methyltetrahydrofolate Homocysteine
Adenosyl
homocysteine hydrolase
Homocysteine

Cystathionine -synthase
Vitamin B6 NH3+

Cystathionine
-OOCCHCH2CH2 SH
-cystathionase Homocysteine
Vitamin B6
Cysteine

Figure 42-13 Homocysteine metabolic pathway. Dietary methionine is converted to homocysteine. Homocysteine is remethylated via methionine synthase
in the presence of vitamin B12 to form metabolic methionine, identical to dietary methionine. This reaction requires 5-methyltetrahydrofolate, which is supplied
through dietary folate, and methylenetetrahydrofolate reductase. Homocysteine is also metabolized via cystathionine -synthase and -cystathionase to cys-
teine, which is excreted in urine or reused in protein metabolism. Cysteine production requires vitamin B6. Deficiencies of vitamin B6, vitamin B12, or folate
and mutations in methionine synthase, methylenetetrahydrofolate reductase, or cystathionine -synthase result in hyperhomocysteinemia.

TABLE 42-11 Homocysteine Enzyme Immunoassay TABLE 42-12 Relative Risk of Coronary Events
Reference Intervals According to Concentration of Fibrinogen*
Population Reference Interval (mol/L) Fibrinogen Concentration
Quintile Relative Risk of Coronary Event
Females 60yr 4.6-12.1
Females >60yr 6.6-14.1 1 1.0
Males 60yr 5.0-15.6 2 1.89
Males >60yr 7.0-17.6 3 2.33
4 2.56
5 2.89
in January 1999 to prevent fetal neural tube defects, has
*The relative risks are shown for each of five quintiles of subjects defined according
reduced the necessity for supplemental vitamin therapy. to the concentrations of each fibrinogen from 1, the group with the lowest concentra-
tion, to 5, the group with the highest. Relative risks have been adjusted for all con-
founding factors. The group with the lowest values serves as the reference group.
Fibrinogen Activity
Fibrinogen may be measured using the clot-based method of
Clauss and predicts cardiovascular risk.82 Fibrinogen concen- Elevated fibrinogen makes blood more viscous, which
trations have a positive correlation with relative risk of myo- favors coagulation, platelet activation, and formation of ath-
cardial infarction in patients with angina pectoris, as shown in erothrombotic lesions. It directly promotes platelet activation
Table 42-12. The relative risk triples from the first to the fifth by binding to their glycoprotein IIb/IIIa membrane receptors.
quintiles, and even high-normal levels predict increased risk. Because fibrinogen becomes integrated into atherothrombotic
This predictive capacity is seen in both healthy people and lesions, it contributes to their thrombotic potential.
patients with angina.83 There is a relationship between fibrino- Although fibrinogen is a strong predictor of arterial throm-
gen and total cholesterol levels (Figure 42-14). High fibrinogen bosis, the use of the fibrinogen assay for this purpose is limited.
concentrations can be used to predict hypercholesterolemia There are no independent therapeutic regimens that specifically
and identify patients who are at high risk for new coronary lower fibrinogen, and no clinical trials suggest that reduction of
events. In contrast, low fibrinogen levels are associated with fibrinogen concentrations in blood reverses the odds of throm-
low risk of cardiovascular events, even in patients with high bosis. Further, fibrinogen assays are not standardized. Clinical
total cholesterol levels. hemostasis laboratories may use the clot-based Clauss assay,
CHAPTER 42 Thrombosis Risk Testing 683

systems: vascular intima, platelets, leukocytes, coagulation,


coagulation control pathways, and fibrinolysis.85 In DIC, fibrin
6.9
microthrombi partially occlude small vessels and lead to
7.0 5.3 consumption of platelets, coagulation factors, coagulation
6.0 control proteins, and fibrinolytic enzymes. Fibrin degradation
Risk of coronary events

5.0 4.1 products (FDPs, traditionally called fibrin split products, FSPs)
3.6 become elevated and interfere with normal fibrin formation.86
4.0
3.5 3.4 Many toxic and inflammatory processes are set loose.87
3.0
1.2 DIC may be acute and uncompensated, with deficiencies of
1.6
2.0 multiple hemostasis components, or chronic, with normal or
1.3 High even elevated clotting factor levels. In chronic DIC, liver coagu-
1.0
Middle lation factor production and bone marrow platelet production
Cholesterol
0.0 Low
compensate for increased consumption.
High

Middle

Low

Although DIC is a thrombotic process, the thrombi that


form are small and ineffective, so systemic hemorrhage is
Fibrinogen
often the first or most apparent symptom. Acute DIC is
Figure 42-14 Coronary risk prediction by fibrinogen and cholesterol con- often fatal and requires immediate medical intervention. The
centrations. Fibrinogen and total cholesterol synergistically predict coronary diagnosis relies heavily on the hemostasis laboratory, and
risk. Tertiles of fibrinogen concentration are shown in relation to tertiles of
medical laboratory scientists often perform a DIC laboratory
total cholesterol concentration with the relative risk of coronary events indi-
profile under medical emergency circumstances. Defibrination
cated for each combination.
syndrome and consumption coagulopathy are alternate names
for DIC.
TABLE 42-13 Normal Ranges for Lipoprotein (a) by
Race and Sex Causes
Any disorder that contributes hemostatic molecules or pro-
Race Males Females
motes their endogenous secretion may cause DIC. Classifying
African American 4.7-71.8mg/dL 4.4-75.0mg/dL and listing all the causes of DIC is impossible, but the major
European American 2.2-49.4mg/dL 2.1-57.3mg/dL triggering mechanisms and examples of each are listed in
Table 42-14.
The more acutely ill the patient, the more dangerous the
immunoassay, or nephelometry, all yielding varying results. symptoms. Chronic DIC may be associated with vascular
Nevertheless, statin (cholesterol-lowering) therapy, smoking tumors, tissue necrosis, liver disease, renal disease, chronic
cessation, and exercise lower fibrinogen levels along with inflammation, use of prosthetic devices, and adenocarcinoma.
LDL-C and total cholesterol levels, and the assay results parallel The malignancies most associated with DIC are pancreatic,
those for the other members of the risk prediction profile. prostatic, ovarian, and lung cancers; multiple myeloma; and
myeloproliferative diseases. Acute DIC is seen in association
Lipoprotein (a) with obstetric emergencies, intravascular hemolysis, septice-
Lipoprotein (a) is an LDL with noteworthy thrombosis risk mia, viremia, burns, acute inflammation, crush injuries, dis-
prediction characteristics. The plasma level is measured by secting aortic aneurysms, and cardiac disorders.
enzyme immunoassay, and its reference ranges are shown in
Table 42-13. Although lipoprotein (a) concentrations are Pathophysiology
higher in African Americans than in European Americans, the Triggering events may activate coagulation at any point in
level is a stronger predictor of thrombosis in whites, and its the pathway. When triggered, however, DIC proceeds in a
measurement should be considered to be a part of an arterial predictable sequence of events. Circulating thrombin is the
thrombosis prediction profile.84 primary culprit, because it activates platelets, catalyzes fibrin
Lipoprotein (a) may contribute to thrombosis by its antifi- formation, and consumes control proteins. The fibrinolytic
brinolytic property. The molecule competes with plasminogen system may become activated at the level of plasminogen or
for binding sites on newly formed fibrin polymer, which TPA subsequent to fibrin formation, and endothelial cells may
decreases the plasmin activity available for clot degradation. It become damaged, releasing coagulation active substances.
also may contribute to the overall concentration of LDL-C. Finally, leukocytes, particularly monocytes, may be induced
Levels may be lowered with statin drugs, as are LDL-C levels. to secrete tissue factor by the cytokines released during
inflammation.
Thrombin cleaves fibrinogen, creating fibrin monomers. In
DISSEMINATED INTRAVASCULAR
normal hemostasis, fibrin monomers spontaneously polymer-
COAGULATION
ize to form an insoluble gel. The polymer becomes strength-
DIC is a generalized activation of hemostasis secondary to a ened through cross-linking, binding plasminogen as it forms.
systemic disease and is often fatal. DIC involves all hemostatic In DIC, a percentage of fibrin monomers fail to polymerize and
684 PART VIII Hemostasis and Thrombosis

TABLE 42-14 Conditions Associated with Disseminated Intravascular Coagulation Grouped by Mechanism
Mechanism Examples of Conditions
Release of tissue factor into circulation through endothelial cell damage or Physical trauma: crush or brain injuries, surgery
monocyte activation Degradation of muscle; rhabdomyolysis
Tissue ischemia; myocardial infarction
Thermal injuries: burns or cold
Adenocarcinoma
Exposure of subendothelial tissue factor during vasodilatation Hypovolemic and hemorrhagic shock
Malignant hypertension
Asphyxia and hypoxia
Heat stroke
Vasculitis
Endotoxins that activate cytokines Bacterial, protozoal, fungal, and viral infections
Toxic shock syndrome
Circulating immune complexes Heparin-induced thrombocytopenia with thrombosis
Use of drugs that trigger an immune response
Acute hemolytic transfusion reactions
Allergic reactions and anaphylaxis
Bacterial and viral infections
Graft rejection
Particulate matter from tissue injury Eclampsia, preeclampsia, HELLP syndrome
Retained dead fetus or missed abortion
Amniotic fluid embolism
Abruptio placentae
Rupture of uterus
Tubal pregnancy
Fat embolism
Heat stroke
Infusion of activated clotting factors Activated prothrombin complex concentrate therapy
Secretion of proteolytic enzymes Acute promyelocytic or myelomonocytic leukemia
Bacterial, protozoal, fungal, and viral infections
Pancreatitis
Toxins that trigger coagulation Snake or spider envenomation
Pancreatitis
Thrombotic disease or thrombogenic conditions Thrombotic thrombocytopenic purpura, hemolytic uremic syndrome
Pregnancy, postpartum period, estrogen therapy
Deep vein thrombosis, pulmonary embolus
Coagulation control system deficiencies
Purpura fulminans, skin necrosis
Severe hypoxia or acidosis Acute coagulopathy of trauma-shock
Chronic inflammation
Diabetes mellitus
Platelet activation Vascular surgery, coronary artery bypass surgery
Thrombocytosis, thrombocythemia, polycythemia
Vascular tumors
Vascular prostheses
Aortic aneurysm

HELLP, Hemolysis, elevated liver enzymes, and low platelet count.

circulate in plasma as soluble fibrin monomers. The monomers fibrin clot to which it is bound (see Chapter 40). In DIC,
coat platelets and coagulation proteins, creating an anticoagu- plasmin circulates in the plasma and digests all forms of
lant effect (Figure 42-15). fibrin.88 Consequently, fibrin degradation products labeled X,
Soluble fibrin monomers, fibrin polymer, and cross-linked Y, D, E, and D-dimer are detectable in the plasma in concentra-
fibrin all activate plasminogen. Normally, the active form of tions exceeding 20,000ng/mL (Figure 42-16). D-dimer arises
plasminogen, plasmin, acts locally to digest only the solid from cross-linked fibrin polymer, whereas the other fibrin
CHAPTER 42 Thrombosis Risk Testing 685

Thrombin
D domain D domain

A
B

A
A
B E domain
B
A

A A
B B

Fibrinogen D E D
Thrombin cleaves
fibrinopeptides A and B
Fibrin monomer D E D

Spontaneous polymerization

D E D
Fibrin polymer
D E D D E D
XIIIa cross-links D domains
Stabilized fibrin D E D D E D
D E D D E D D E D
D E D D E D
B
Figure 42-15 Normal fibrinogen polymerization. A, Thrombin cleaves fibrinopeptides A and B from the and chains of fibrinogen, creating fibrin monomer
with D and E domains. B, Fibrin monomer normally polymerizes to form long polymers. These are then cross-linked by the action of factor XIIIa. When excess
fibrin degradation products are present, the fibrin monomer fails to polymerize normally, which causes an increase in plasma fibrin monomer.

Thrombin
FXllla

Fibrinogen Fibrin polymer, cross-linked fibrin X, Y, D, E, and D-D

TPA Bound TAFI


plasmin

Plasminogen

Free plasmin TPA PAI-1

2
antiplasmin

Figure 42-16 Fibrinolysis components and controls. Fibrin polymer and cross-linked fibrin are formed by the enzymatic action of thrombin (activated
coagulation factor II, or IIa) and activated coagulation factor XIII (FXIIIa), respectively, on plasma fibrinogen. Plasma plasminogen becomes bound to fibrin
adjacent to tissue plasminogen activator (TPA). Over a period of hours, bound TPA converts bound adjacent plasminogen to plasmin, which digests fibrin to
form fibrin degradation products (FDPs) X, Y, D, E, and D-dimer (D-D). The D-D is produced from cross-linked fibrin. Free plasmin is neutralized by 2-
antiplasmin and TPA is neutralized by plasminogen activator inhibitor 1 (PAI-1). Bound plasmin is controlled by thrombin-activatable fibrinolysis inhibitor (TAFI).

degradation products may be produced from fibrinogen or is lost as protein C, protein S, and antithrombin are consumed.
fibrin monomers or polymers.89 The combination of thrombin activation, circulating plasmin,
Platelets become enmeshed in the fibrin polymer or are loss of control, and thrombocytopenia contribute to the overall
exposed to thrombin; both events trigger platelet activation, hemorrhagic outcome of DIC.
which drives the coagulation system and produces thrombocy- Plasmin digests factors V, VIII, IX, and XI as well as other
topenia. At the same time, control of the coagulation pathway plasma proteins. Plasmin also may trigger complement, which
686 PART VIII Hemostasis and Thrombosis

leads to hemolysis, and the kinin system, which results in venous thrombosis such as deep vein thrombosis or pulmo-
inflammation, hypotension, and shock. During fibrinolytic nary emboli, in which levels increase to greater than
therapy, in amyloidosis, and sometimes in liver disease, 500ng/mL.93 D-dimer concentrations are typically elevated in
plasminogen is activated independently of the coagulation inflammation, sickle cell crisis, pregnancy, and renal disease,
pathway. This condition, called systemic fibrinolysis, produces so an abnormal result alone cannot be used to diagnose
laboratory-measurable fibrin degradation products, including venous thromboembolism definitively.94 The judicious use of
D-dimer, and prolonged prothrombin time (PT) and PTT with the D-dimer assay reduces the requirement for invasive diag-
a normal platelet count.90 nostic tests such as pulmonary angiography when pulmonary
embolism is suspected.95
Symptoms
The symptoms signaling DIC frequently are masked by the Specialized Laboratory Tests That May Aid
symptoms of the underlying disorder and may be chronic, in Diagnosis
acute, or fulminant. Thrombosis in the microvasculature of Table 42-16 lists specialized laboratory tests that may be used
major organs may produce symptoms of organ failure, such as to diagnose and classify DIC in special circumstances. Results
renal function impairment, adult respiratory distress syn- consistent with DIC and clinical comments are included.
drome, and central nervous system manifestations. Skin, bone, Many of these tests are available in acute care facilities, but are
and bone marrow necrosis may be seen. Purpura fulminans not routinely applied to diagnosis of DIC. Others are available
is seen in meningococcemia, chickenpox, and spirochete only in tertiary care facilities and hemostasis reference
infections. laboratories.96-98
Protein C, protein S, and antithrombin typically are con-
Laboratory Diagnosis sumed in DIC, and their assay adds little to the diagnosis;
DIC is a clinical diagnosis that requires laboratory confirma- however, when thawed frozen plasma, APC (Xigris; Eli Lilly,
tion (see Chapter 45). The primary profile includes a platelet Indianapolis, Ind.), or antithrombin concentrate (Thrombate
count, blood film examination, PT, PTT, D-dimer, and fibrino- III) is used to treat DIC, these assays are useful in establishing
gen assays. Anticipated results in DIC are shown in Table the necessity for therapy and monitoring its effect. Factor assays
42-15. Prolonged PT and PTT reflect coagulation factor con- may clarify PT and PTT results. The thrombin time and the
sumption. Fibrinogen concentrations may decrease in DIC, but reptilase time also are sensitive DIC screens.
because fibrinogen is an acute phase reactant that increases in Tests of the fibrinolytic pathway include serum fibrin deg-
inflammation, the fibrinogen concentration alone provides radation products, chromogenic plasminogen activity assay,
little information and may exceed 400mg/dL. The peripheral TPA and PAI-1 immunoassays, and the euglobulin clot lysis
blood film platelet estimate confirms thrombocytopenia in time test, a time-honored clot-based assay that offers only
nearly all cases, and the presence of schistocytes helps establish modest specificity.99 These tests are seldom offered in the acute
the diagnosis of DIC in about 50% of cases.91,92 An elevated care hemostasis laboratory because they require careful speci-
D-dimer level is essential to the diagnosis. D-dimer also may men management, but they may help in the diagnosis of
be elevated in inflammatory conditions, localized thrombosis, primary fibrinolysis, sometimes seen after fibrinolytic therapy.
or renal disease in the absence of DIC; the PT, PTT, platelet
count, blood film examination, and other laboratory assays Treatment
must be performed with the D-dimer to rule out these To arrest DIC, the physician must diagnose and treat the under-
disorders. lying disorder. Surgery, antiinflammatory agents, antibiotics,
The D-dimer reference limit is typically 240ng/mL, although or obstetric procedures as appropriate may normalize hemo-
the interval varies with location and technology. In 85% of DIC stasis, particularly in chronic DIC. Supportive therapy, such as
cases, D-dimer levels reach 10,000 to 20,000ng/mL. A normal maintenance of nutrition and fluid and electrolyte balance,
D-dimer assay result rules out DIC and rules out localized always accompanies the management.
In acute DIC, in which multiorgan failure from micro-
thrombosis and bleeding threatens the life of the patient,
TABLE 42-15 Anticipated Results of Disseminated
heroic measures are necessary. Treatment falls into two catego-
Intravascular Coagulation (DIC) Primary Laboratory Profile
ries: therapies that slow the clotting process and therapies that
Reference replace missing platelets and coagulation factors.
Test Interval Value in DIC Heparin may be used for its antithrombotic properties to
Platelet count 150-450/mcL <150/mcL stop the uncontrolled activation of the coagulation cascade.
Prothrombin time 11-14sec >14sec Because heparin may aggravate bleeding, careful observation
Partial thromboplastin 25-35sec >35sec and support are required. Repeated chromogenic anti-Xa
time heparin assays may be necessary to control heparin dosage,
D-dimer 0-240ng/mL >240ng/mL, often 10,000 because in DIC the PTT is ineffective for monitoring heparin
to 20,000ng/mL therapy.
Fibrinogen 220-498mg/dL <220mg/dL Thawed frozen plasma provides all the necessary coagula-
tion factors and replaces blood volume lost during acute DIC
CHAPTER 42 Thrombosis Risk Testing 687

TABLE 42-16 Specialized Hemostasis Laboratory Assays Useful in Diagnosis and Classification of Disseminated
Intravascular Coagulation (DIC)
Assay Value in DIC Comments
Serum FDP >10mcg/mL Obsolete, replaced by quantitative D-dimer.
Soluble fibrin monomer Positive Hemagglutination assay provides valid measure of fibrin monomer. Avoid obsolete
tests such as protamine sulfate solubility or ethanol gelation.
Thrombus precursor protein >3.5mcg/mL Immunoassay with no interference from fibrinogen or FDPs.
Protein C, protein S, and AT activity assays <50% Use to monitor therapy: thawed frozen plasma, AT concentrate, and activated
protein C concentrate.
Factor assays: II (prothrombin), V, VIII, X <30% Factors V and VIII rise in inflammation, and thus assays may give misleading results.
Thrombin time, reptilase time Prolonged Fibrinogen levels <80mg/dL, elevated FDPs, and soluble fibrin monomer all prolong
thrombin time and reptilase time.
Plasminogen, tissue plasminogen activator Decreased May be useful for further analyzing systemic fibrinolysis. Specimen management
protocol must be strictly observed.
Euglobulin clot lysis time <2hr or >12hr Endpoint is clot dissolution. Shortened lysis time indicates increased fibrinolysis,
prolonged lysis time indicates deficiency; both may occur in DIC.
Peripheral blood film exam Anemia with Schistocytes (microangiopathic hemolytic anemia) are present in 50% of DIC cases;
schistocytes leukocytosis is common.
Localized thrombosis markers: prothrombin Elevated Most useful in diagnosis of localized thrombotic events but may be used to monitor
fragment 1+2, thrombin-antithrombin DIC therapy. Used in clinical trials.

AT, Antithrombin; FDP, fibrin degradation product.

hemorrhage. Its effects are best monitored with serial PT and The TAT covalent complex is formed when antithrombin
PTT. Prothrombin complex concentrate (Proplex T complex; neutralizes thrombin. This reaction is enhanced by the presence
Baxter Healthcare Corporation, Deerfield, Ill.) is rarely needed, of heparin. TAT has a half-life of 3 minutes and a normal range
but may be an alternative if plasma expansion must be avoided. of 0.5 to 5ng/mL. To avoid in vitro release or activation of PF
Fibrinogen is often of paramount concern, and cryoprecipitate 1+2 or TAT, plasma specimens are collected in 3.2% sodium
supplies it at high concentrations and low volume, along with citrate and are centrifuged and separated within minutes of
concentrated factor VIII. Repeated measurements of fibrinogen, collection, and the plasma is frozen until ready for assay.
PT, and PTT are necessary to confirm the effectiveness of cryo-
precipitate administration. Platelet transfusions are necessary
HEPARIN-INDUCED THROMBOCYTOPENIA
if thrombocytopenia is severe. The effectiveness of platelet con-
centrate as well as platelet consumption are monitored with Heparin-induced thrombocytopenia (HIT), also called heparin-
platelet counts (see Chapter 45). Red blood cells are adminis- induced thrombocytopenia with thrombosis, is an adverse effect of
tered as necessary to treat the resulting anemia. Antifibrinolytic treatment with unfractionated heparin.
therapy is contraindicated except in proven systemic
fibrinolysis.100 Cause and Clinical Significance
Between 1% and 5% of patients receiving unfractionated
heparin for more than 5 days develop an IgG antibody to
LOCALIZED THROMBOSIS MONITORS
heparinplatelet factor 4 immune complexes. In 30% to 50%
In addition to D-dimer, several peptides and coagulation factor of cases, the immune complexes that are formed bind platelet
complexes are released into the plasma during coagulation. Fc receptors, which leads to platelet activation, thrombocyto-
One complex, TAT, and one peptide, PF 1+2, may be assayed penia, and formation of microvascular thrombi.103,104 HIT
as a means to detect and monitor localized venous or arterial occurs with intravenous and subcutaneous heparin administra-
thromboses. TAT and PF 1+2 immunoassays are sensitive but tion at prophylactic and therapeutic dosages, although it is
nonspecific, because levels become elevated not only in throm- more frequent with therapeutic doses.105 Venous thrombosis
bosis but also in DIC, septicemia, eclampsia, pancreatitis, predominates 5:1, but arterial thrombosis accounts for the
leukemia, liver disease, and trauma.101,102 These assays are of most disturbing symptoms. Patients may develop pulmonary
particular value in clinical trials of anticoagulants. emboli, limb gangrene requiring amputation, stroke, and myo-
PF 1+2 is released from prothrombin at the time of its con- cardial infarction. HIT is often a medical emergency, and the
version to thrombin by the prothrombinase complex. It has a mortality rate is 20%.106
plasma half-life of 90 minutes and a reference range of 0.3 to
1.5nmol/L. Elevated PF 1+2 may be seen in venous thrombo- Platelet Count
embolism. Heparin or oral anticoagulant therapy reduces its Patients receiving heparin must have platelet counts performed
plasma concentration. every other day. A decrease in platelet count during heparin
688 PART VIII Hemostasis and Thrombosis

TABLE 42-17 The 4Ts Scoring System: Laboratory and Clinical Pretest Probability of Heparin-Induced
Thrombocytopenia (HIT)
SCORE
Indicator 2 1 0
Acute Thrombocytopenia >50% decrease in platelet count to 30% to 50% decrease in platelet count, or <30% decrease in
nadir of 20,000/mcL >50% if directly resulting from surgery, platelet count, or to
to nadir of 10,000 to 19,000/mcL nadir of <10,000/mcL
Timing of platelet count decrease, Onset of decrease on day 5 to 10, Apparent decrease on day 5 to 10, but Decrease at 4 days
thrombosis, or other sequelae or onset of decrease on day 1 if unclear due to missing platelet counts; without recent heparin
of HIT (first day of heparin previous heparin exposure within decrease after day 10; decrease on day exposure
therapy is day 0) past 5 to 30 days 1 if previous heparin exposure within
past 31 to 100 days
Thrombosis, skin lesions, acute Proven new thrombosis or skin Progressive, recurrent, or suspected None
system reaction necrosis; acute systemic reaction thrombosis; erythematous skin lesions
after intravenous heparin bolus
OTher causes for No explanation for platelet count Possible other cause is evident Definite other cause is
thrombocytopenia decrease is evident evident

From Crowther MA, Cook DJ, Albert M, et al: Canadian Critical Care Trials Group: the 4Ts scoring system for heparin-induced thrombocytopenia in medical-surgical intensive
care unit patients, J Crit Care 25:287-293, 2010.
Maximum pretest probability score is 8; a score of 6 to 8 indicates high probability of HIT; 4 to 5, intermediate probability; 0 to 3, low probability.
The most immunizing heparin exposure is considered first; unfractionated heparin (UFH) received during cardiac surgery is more immunogenic than UFH or low-molecular-weight
heparin received for acute coronary syndrome. The day the platelet count begins to fall is considered the day of onset of thrombocytopenia. It generally takes 1 to 3 days before
an arbitrary threshold that defines thrombocytopenia, such as 150,000 platelets/mcL, is passed.

administration is a recognized indicator of HIT, but the inter- technically demanding and is only 50% sensitive for HIT. Also
pretation of the thrombocytopenia is confounded, because technically demanding, but perhaps more sensitive, is washed
30% of patients receiving heparin develop an immediate, platelet suspension lumiaggregometry. The reference method
benign, and limited thrombocytopenia, sometimes called HIT is the carbon 14 serotonin washed platelet release assay,
type I.107 This benign form of thrombocytopenia usually devel- provided by reference laboratories that possess radionuclide
ops in 1 to 3 days, whereas immune-mediated HIT, sometimes licenses.
called HIT type II, develops after 5 days. There is significant
overlap in patients previously exposed to heparin. In HIT (HIT Treatment
type II), the decrease in platelet count may exceed 40%, When heparin-induced antibodies are detected, the adminis-
whereas in benign thrombocytopenia the decrease is relatively tration of heparin must be discontinued immediately, and
small; however, in both cases, the platelet count may remain an alternate form of anticoagulation must be considered.
within the normal range. The HIT diagnosis is made using the Complete cessation of anticoagulant therapy is risky, because
4Ts approach, shown in Table 42-17. additional thrombotic events are likely to occur. Although
low-molecular-weight heparin causes HIT in fewer than 1% of
Other Laboratory Tests cases in which it is the sole anticoagulant, its use is contrain-
Because at least 10% of hospitalized patients receive heparin, dicated when HIT already has been induced during unfraction-
the acute care laboratory must provide a procedure to confirm ated heparin therapy. Likewise, oral anticoagulant therapy is
HIT and differentiate it from benign thrombocytopenia. The discouraged, because this may precipitate a potentially fatal
heparin-induced antibody immunoassay is available in most warfarin-induced skin necrosis syndrome. Synthetic penta
acute care facility laboratories, although seldom as a stat assay saccharide, which mimics heparins antithrombin-binding
(see Chapter 45).108 The assay result may become positive sequence, appears safe to use in HIT.
before clinical signs of HIT are evident and is more sensitive Recombinant lepirudin (ZLB Behring GmbH, Marburg,
than aggregometry. The result may be negative in a few HIT Germany) is a direct thrombin inhibitor modeled after leech
patients, however, possibly because peptides other than plate- saliva that binds the reactive site of thrombin. Lepirudin is
let factor 4 also may form complexes with heparin. Microbial administered as a continuous infusion, and therapy may
contamination, lipemia, and hemolysis may also invalidate the be monitored using the PTT or the Ecarin prothrombin
test results. The presence of immune complexes or immuno- assay (Pentapharm, Basel, Switzerland) based on reagent
globulin aggregates in the patient specimen may cause increased derived from Echis carinatus snake venom, available only from
nonspecific binding and produce false-positive results. reference laboratories. Argatroban, another direct thrombin
Many laboratories provide aggregometry or lumiaggregome- inhibitor, also may be used, and therapy may be monitored
try for the diagnosis of HIT (Chapter 45).109 Aggregometry is using the PTT.
CHAPTER 42 Thrombosis Risk Testing 689

current area of development is the prediction of risk of cardio-


CONCLUSION
vascular and cerebrovascular disease. The future may give us
The importance of laboratory diagnosis has become evident in markers of endothelial cell disease, measures of leukocyte
all forms of thrombotic diseasechronic and acute, arterial adhesion, and more specific markers of inflammation and
and venous, primary and secondary. The most promising platelet activity.

SUMMARY
Thrombosis is the most prevalent condition in developed countries The tests for evaluating the protein C pathway include protein C
and accounts for most illness and premature death. and protein S activity and concentration, APC resistance, FVL
Thrombosis may be arterial, causing peripheral artery disease, assay, and C4bBP assay.
heart disease, and stroke, or venous, causing deep vein thrombo- The molecular test for the prothrombin G20210A mutation predicts
sis and pulmonary emboli. the risk of venous thrombosis.
Most thrombosis occurs as a result of lifestyle habits and aging, DIC is a clinical diagnosis confirmed by a series of assays in the
but many thrombotic disorders are related to congenital risk acute care facility.
factors. Chronic thrombosis may be identified using the PF 1+2, TAT
Thrombosis risk profiles may be offered to clinicians for screening complex, and quantitative D-dimer assays.
purposes in high-risk populations. The laboratory provides confirmatory tests in HIT with
The main hemostasis predictors of arterial thrombotic disease are thrombosis.
elevated levels of CRP (measured by high-sensitivity assay),
homocysteine, fibrinogen, lipoprotein (a), and coagulation factors. Now that you have completed this chapter, go back and
APL antibody testing requires a series of essential hemostasis read again the case study at the beginning and respond
laboratory assaysclot-based tests and immunoassays. to the questions presented.
Antithrombin may be assayed using chromogenic substrate and
enzyme immunoassay analyses.

R E V I E W Q UESTIONS
1. What is the prevalence of venous thrombosis in the United 5. What is the most common heritable thrombosis risk factor
States? in whites?
a. 0.001 a. Prothrombin G20210A mutation
b. 0.01 b. APC resistance
c. 10% to 15% c. Antithrombin deficiency
d. 500,000 cases per year d. Protein S deficiency

2. What is thrombophilia? 6. In most LA profiles, what test is generally the first used to
a. Predisposition to thrombosis secondary to a screen for LA?
congenital or acquired disorder a. DRVVT
b. Inappropriate triggering of the plasma coagulation b. Low-phospholipid PTT
system c. KCT
c. A condition in which clots form uncontrollably d. PT
d. Inadequate fibrinolysis
7. A patient with venous thrombosis is tested for protein S
3. What acquired thrombosis risk factor is assessed in the deficiency. The protein S activity, antigen, and free antigen
hemostasis laboratory? all are less than 65%, and the C4bBP level is normal. What
a. Smoking type of deficiency is likely?
b. Immobilization a. Type I
c. Body mass index b. Type II
d. LA c. Type III
d. No deficiency is indicated, because the reference range
4. Trousseau syndrome, a low-grade chronic DIC, is often includes 25%
associated with what type of disorder?
a. Renal disease
b. Hepatic disease
c. Adenocarcinoma
d. Chronic inflammation
690 PART VIII Hemostasis and Thrombosis

8. An elevated level of what fibrinolytic system assay is asso 12. What therapeutic agent may occasionally cause DIC?
ciated with arterial thrombotic risk? a. Factor VIII
a. PAI-1 b. Factor VIIa
b. Lipoprotein (a) c. Antithrombin concentrate
c. Factor VIIa d. Activated prothrombin complex concentrate
d. Factor XII
13. Name an old fibrinolysis assay that has been largely
9. How does lipoprotein (a) cause thrombosis? replaced by the quantitative D-dimer assay.
a. It causes elevated factor VIII levels. a. Euglobulin lysis
b. It coats the endothelial lining of arteries. b. Soluble fibrin monomer
c. It contributes additional phospholipid in vivo for c. 5-M urea solubility assay
formation of the Xase complex. d. Fibrinogen degradation products
d. It substitutes for plasminogen or TPA in the forming
clot. 14. What is the most important application of the quantitative
D-dimer test?
10. What test may be used to confirm the presence of LA? a. Diagnose primary fibrinolysis
a. Bethesda titer b. Diagnose liver and renal disease
b. PT c. Rule out deep venous thrombosis
c. Antinuclear antibody d. Diagnose acute myocardial infarction
d. PTT using high-phospholipid reagent

11. What deoxyribonucleic acid (DNA)based test may be


used to confirm APC resistance?
a. Prothrombin G20210A
b. FVL
c. MTHFR 1298
d. MTHFR 677

REFERENCES
1. Francis JL: Laboratory investigation of hypercoagulability. 10. Stein PD, Matta F, Musani MH, et al: Silent pulmonary
Semin Thromb Hemost 24:111-126, 1998. embolism in patients with deep venous thrombosis: a sys-
2. Dahlback B, Carlsson M, Svensson PJ: Familial thrombo- tematic review. Am J Med 123:426-431, 2010.
philia due to a previously unrecognized mechanism charac- 11. Rosendaal FR, Reitsma PH: Genetics of venous thrombosis.
terized by poor anticoagulant response to activated protein J Thromb Haemost 7(suppl 1):301-304, 2009.
C: prediction of a cofactor to activated protein C. Proc Natl 12. Cushman M, Alving BM: Risk factors for cardiovascular
Acad Sci U S A 90:1004-1008, 1993. disease and arterial thrombosis. In Kitchens CS, Alving BM,
3. Bertina RM, Koeleman BPC, Koster T, et al: Mutation in Kessler CM, editors: Consultative hemostasis and thrombosis,
blood coagulation factor V associated with resistance to ed 2, Philadelphia, 2007, Saunders, pp 371-378.
activated protein C. Nature 369:64-67, 1994. 13. Heit JA: Epidemiology of venous thromboembolism. In
4. Nieuwdorp M, Stroes ES, Meijers JC, et al: Hypercoagulabil- Colman RW, Marder VJ, Clowes AW, et al, editors: Hemosta-
ity in the metabolic syndrome. Curr Opin Pharmacol 5:155- sis and thrombosis: basic principles and clinical practice,
159, 2005. ed 5, Philadelphia, 2006, Lippincott Williams & Wilkins,
5. Heit JA: Thrombophilia: clinical and laboratory assessment pp 1227-1234.
and management. In Kitchens CS, Alving BM, Kessler 14. Rosenberg RD, Bauer KA: New insights into hypercoagula-
CM, editors: Consultative hemostasis and thrombosis, ed 2, ble states. Hosp Pract 21:131-138, 1986.
Philadelphia, 2007, Saunders, pp 213-244. 15. Rak J, Milsom C, May L, et al: Tissue factor in cancer and
6. Heart disease and stroke statistics2010 update: a report angiogenesis: the molecular link between genetic tumor
from the American Heart Association statistics committee progression, tumor neovascularization, and cancer coagu-
and stroke statistics subcommittee. Circulation 121:e46- lopathy. Semin Thromb Hemost 32:54-70, 2006.
e215, 2010. 16. De Stefano V, Sora F, Rossi E, et al: The risk of thrombosis
7. Goldhaber SZ: Pulmonary embolism. N Engl J Med 339:93- in patients with acute leukemia: occurrence of thrombosis
104, 1998. at diagnosis and during treatment. J Thromb Haemost 3:1985-
8. Anderson FA, Wheeler HB, Goldberg RJ, et al: A population- 1992, 2005.
based perspective of the hospital incidence and case-fatality 17. Rosse WF, Ware RE: The molecular basis of paroxysmal noc-
rate of deep-vein thrombosis and pulmonary embolism. turnal hemoglobinuria. Blood 86:3277-3286, 1995.
The Worcester DVT study. Arch Intern Med 151:933-938, 18. Garg N, Kumar A, Flaker GC: Antiplatelet therapy for stroke
1991. prevention in atrial fibrillation. Mo Med 107:44-47, 2010.
9. Jensen R: The interface of the physician and the laboratory 19. Bramham K, Hunt BJ, Goldsmith D: Thrombophilia of
in the detection of venous thrombotic riskpart I. Clin nephrotic syndrome in adults. Clin Adv Hematol Oncol
Hemost Rev 11:1-4, 1997. 7:368-372, 2009.
CHAPTER 42 Thrombosis Risk Testing 691
20. Lane DA, Mannuci PM, Bauer KA, et al: Inherited thrombo- hexagonal (II) phase phospholipids. Thromb Haemost 62:
philia: part 2. Thromb Haemost 76:824-834, 1996. 892-896, 1989.
21. Johnson CM, Mureebe L, Silver D: Hypercoagulable states: 42. Palomo I, Segovia F, Ortega C, et al: Antiphospholipid syn-
a review. Vasc Endovasc Surg 39:123-133, 2005. drome: a comprehensive review of a complex and multisys-
22. Itakkura H: Racial disparities in risk factors for thrombosis. temic disease. Clin Exp Rheumatol 27:668-677, 2009.
Curr Opin Hematol 12:364-369, 2005. 43. Tebo AE, Jaskowski TD, Phansalkar AR, et al: Diagnostic
23. Brown K, Luddington R, Williamson D, et al: Risk of venous performance of phospholipid-specific assays for the evalua-
thromboembolism associated with a G to A transition at tion of antiphospholipid syndrome. Am J Clin Pathol
position 20210 in the 3-untranslated region of the pro- 129:870-875, 2008.
thrombin gene. Br J Haematol 98:907-909, 1997. 44. Forastiero R, Martinuzzo M, Pombo G, et al: A prospective
24. Ridker PM, Hennekens CH, Selhub J, et al: Interrelation study of antibodies to beta2-glycoprotein I and prothrom-
of hyperhomocysteinemia, factor V Leiden, and risk of bin, and risk of thrombosis. J Thromb Haemost 3:1231-1238,
future venous thromboembolism. Circulation 95:1777- 2005.
1782, 1997. 45. Blank M, Shoenfeld Y: Antiphosphatidylserine antibodies
25. Heit JA, OFallon WM, Petterson TM, et al: Relative impact and reproductive failure. Lupus 13:661-665, 2004.
of risk factors for deep vein thrombosis and pulmonary 46. Samaha M, Trossaert M, Conard J, et al: Prevalence and
embolism: a population-based study. Arch Intern Med patient profile in activated protein C resistance. Am J Clin
162:1245-1248, 2002. Pathol 104:450-454, 1995.
26. Alarcon-Segovia D, Cabral AR: The concept and classifica- 47. Favaloro EJ, McDonald D, Lippi G: Laboratory investigation
tion of antiphospholipid/cofactor syndromes. Lupus 5:364- of thrombophilia: the good, the bad, and the ugly. Semin
367, 1996. Thromb Hemost 35:695-710, 2009.
27. Lim W, Crowther MA, Eikelboom JW: Management of 48. Dahlback B, Hildebrand B: Inherited resistance to activated
antiphospholipid antibody syndrome: a systematic review. protein C is corrected by anticoagulant cofactor activity
JAMA 295:1050-1057, 2006. found to be a property of factor V. Proc Natl Acad Sci U S A
28. Pengo V, Tripodi A, Reber G, et al: Update of the guidelines 91:1396-1400, 1994.
for lupus anticoagulant detection. Subcommittee on Lupus 49. De Ronde H, Bertina RM: Laboratory diagnosis of APC-
Anticoagulant/Antiphospholipid Antibody of the Scientific resistance: a critical evaluation of the test and the develop-
and Standardisation Committee of the International Society ment of diagnostic criteria. Thromb Haemost 72:880-886,
on Thrombosis and Haemostasis. J Thromb Haemost 7:1737- 1994.
1740, 2009. 50. Ragland BD, Reed CE, Eiland BM, et al: The effect of lupus
29. Kandiah DA, Sheng YH, Krilis SA: Beta-2 glycoprotein I: anticoagulant in the second-generation assay for activated
target antigen for autoantibodies in the antiphospholipid protein C resistance. Am J Clin Pathol 119:66-71, 2003.
syndrome. Lupus 5:381-386, 1996. 51. Emadi A, Crim MT, Brotman DJ, et al: Analytic validity of
30. Lie JT: Vasculopathy of the antiphospholipid syndromes genetic tests to identify factor V Leiden and prothrombin
revisited: thrombosis is the culprit and vasculitis the consort. G20210A. Am J Hematol 85:264-270, 2010.
Lupus 5:368-371, 1966. 52. Poort SR, Rosendaal FR, Reitsma PY, et al: A common genetic
31. Lopez-Pedrera C, Buendia P, Barbarroja N, et al: variation in the 3 untranslated region of the prothrombin
Antiphospholipid-mediated thrombosis: interplay between gene is associate with elevated plasma prothrombin levels
anticardiolipin antibodies and vascular cells. Clin Appl and an increase in venous thrombosis. Blood 88:3698-3703,
Thromb Hemost 12:41-45, 2006. 1996.
32. Hughes GRV: The antiphospholipid syndrome. Lupus 5:345- 53. Danckwardt S, Hartmann K, Gehring NH, et al: 3 end pro-
346, 1996. cessing of the prothrombin mRNA in thrombophilia. Acta
33. Rand JH, Levin MB, Alving BM: The antiphospholipid syn- Haematol 115:192-197, 2006.
drome: clinical presentation, diagnosis, and patient man- 54. Naeem MA, Anwar M, Ali W, et al: Prevalence of prothrom-
agement. In Kitchens CS, Alving BM, Kessler CM, editors: bin gene mutation (G-A 20210 A) in general population: a
Consultative hemostasis and thrombosis, ed 2, Philadelphia, pilot study. Clin Appl Thromb Hemost 12:223-226, 2006.
2007, Saunders, pp 319-338. 55. Thaler E, Lechner K: Antithrombin III deficiency and throm-
34. Cohen D, Berger SP, Steup-Beekman GM, et al: Diagnosis boembolism. Clin Haematol 10:369-390, 1981.
and management of the antiphospholipid syndrome. BMJ 56. Wu O, Robertson L, Twaddle S, et al: Screening for throm-
340:c2541, 2010. bophilia in high-risk situations: systematic review and cost-
35. Devreese K, Hoylaerts MF: Challenges in the diagnosis of the effectiveness analysis. The Thrombosis Risk and Economic
antiphospholipid syndrome. Clin Chem 56:930-940, 2010. Assessment of Thrombophilia Screening (TREATS) study.
36. Marques MB, Fritsma GA: Quick guide to coagulation testing, Health Technol Assess 10:1-110, 2006.
ed 2, Washington, DC, 2009, AACC Press. 57. Rosen S: Chromogenic methods in coagulation diagnostics.
37. Kaczor DA, Bickford NN, Triplett DA: Evaluation of different Hamostaseologie 25:259-266, 2005.
mixing study reagents and dilution effect in lupus antico- 58. Antovic J, Sderstrm J, Karlman B, et al: Evaluation of a
agulant testing. Am J Clin Pathol 95:408-411, 1991. new immunoturbidimetric test (Liatest antithrombin III)
38. Sato K, Kagami K, Asai M, et al: Detection of lupus antico- for determination of antithrombin antigen. Clin Lab Hae-
agulant using a modified diluted Russells viper venom test. matol 23:313-316, 2001.
Rinsho Byori 43:263-268, 1995. 59. Spiess BD: Treating heparin resistance with antithrombin or
39. Brandt JT, Triplett DA, Alving B, et al: Criteria for the diag- fresh frozen plasma. Ann Thorac Surg 85:2153-2160, 2008.
nosis of lupus anticoagulants: an update. Thromb Haemost 60. Dahlback B: The protein C anticoagulant system: inherited
74:1185-1190, 1995. defects as basis for venous thrombosis. Thromb Res 77:1-43,
40. Devreese KM: Interpretation of normal plasma mixing 1995.
studies in the laboratory diagnosis of lupus anticoagulants. 61. Heit JA: Epidemiology of venous thromboembolism. In
Thromb Res 119:369-376, 2007. Colman RW, Marder VJ, Clowes AW, et al, editors: Hemosta-
41. Rauch J, Tannenbaum H, Janoff AS: Distinguishing plasma sis and thrombosis: basic principles and clinical practice, ed 5,
lupus anticoagulants from anti-factor antibodies using Philadelphia, 2006, Lippincott William & Wilkins.
692 PART VIII Hemostasis and Thrombosis

62. Mann HJ, Short MA, Schlichting DE: Protein C in critical 84. Deb A, Caplice NM: Lipoprotein(a): new insights into
illness. Am J Health Syst Pharm 15(66):1089-1096, 2009. mechanisms of atherogenesis and thrombosis. Clin Cardiol
63. Desancho MT, Dorff T, Rand JH: Thrombophilia and the risk 27:258-264, 2004.
of thromboembolic events in women on oral contraceptives 85. LaBelle C, Kitchens CS: Disseminated intravascular coagula-
and hormone replacement therapy. Blood Coagul Fibrinolysis tion. In Kitchens CS, Alving MB, Kessler CM, editors: Con-
24:2010, Epub ahead of print. sultative hemostasis and thrombosis, ed 2, Philadelphia, 2007,
64. Khor B, Van Cott EM: Laboratory tests for protein C defi- Saunders, pp 183-198.
ciency. Am J Hematol 85:440-442, 2010. 86. Giles AR: Disseminated intravascular coagulation. In Bloom
65. Van Cott EM, Ledford-Kraemer M, Meijer P, et al: NASCOLA AL, Forbes CD, Thomas DP, et al, editors: Haemostasis
Proficiency Testing Committee. Protein S assays: an analysis and thrombosis, New York, 1994, Churchill Livingstone,
of North American Specialized Coagulation Laboratory pp 969-986.
Association proficiency testing. Am J Clin Pathol 123:778- 87. Marder VJ, Feinstein DI, Colman RW, et al: Consumptive
785, 2005. thrombohemorrhagic disorders. In Colman RW, Marder VJ,
66. Collection, transport, and processing of blood specimens Clowes AW, et al, editors: Hemostasis and thrombosis: basic
for testing plasma-base coagulation assays, approved guide- principles and clinical practice, ed 5, Philadelphia, 2006,
line, ed 4, Document H21-A5, Wayne, Pa, 2008, Clinical Lippincott Williams & Wilkins, pp 1571-1600.
Laboratory and Standards Institute. 88. Vaughan DE, Declerck PJ: Regulation of fibrinolysis. In
67. Ens GE, Newlin F: Spurious protein S deficiency as a result Loscalzo J, Schafer AI, editors: Thrombosis and hemorrhage,
of elevated factor VII levels. Clin Hemost Rev 9:18, 1995. Philadelphia, 2003, Lippincott Williams & Wilkins, pp 105-
68. Morrow DA, Ridker PM: High sensitivity C-reactive protein 120.
(hs-CRP): a novel risk marker in cardiovascular disease. Prev 89. Jensen R: The diagnostic use of fibrin breakdown products.
Cardiol 1:13-16, 1999. Clin Hemost Rev 12:1-2, 1998.
69. Ridker PM: Evaluating novel cardiovascular risk factors: can 90. Wada H, Kobayashi T, Abe Y, et al: Elevated levels of soluble
we better predict heart attacks? Ann Intern Med 130:933-937, fibrin or D-dimer indicate high risk of thrombosis. J Thromb
1999. Haemost 4:1253-1258, 2006.
70. Ridker PM: Fibrinolytic and inflammatory markers for arte- 91. Labelle CA, Kitchens CS: Disseminated intravascular coagu-
rial occlusion: the evolving epidemiology of thrombosis lation: treat the cause, not the lab values. Cleve Clin J Med
and hemostasis. Thromb Haemost 78:53-59, 1997. 72:377-378, 383-385, 390, 2005.
71. Pepys MB, Baltz ML: Acute phase proteins with special refer- 92. Mos IC, Klok FA, Kroft LJ, et al: Safety of ruling out acute
ence to C-reactive protein and related proteins (pentaxins) pulmonary embolism by normal computed tomography
and serum amyloid A proteins. Adv Immunol 34:141-212, pulmonary angiography in patients with an indication for
1983. computed tomography: systematic review and meta-
72. Wilkins J, Gallimore R, Moore E, et al: Rapid automated analysis. J Thromb Haemost 7:1491-1498, 2009.
high sensitivity enzyme immunoassay of C-reactive protein. 93. Janssen MC, Sollersheim H, Verbruggen B, et al: Rapid
Clin Chem 44:1358-1361, 1998. D-dimer assays to exclude deep venous thrombosis and
73. Ridker PM, Cushman M, Stampfer MJ, et al: Inflammation, pulmonary embolism: current status and new develop-
aspirin, and the risk of cardiovascular disease in apparently ments. Semin Thromb Hemost 24:393-400, 1998.
healthy men. N Engl J Med 336:973-979, 1997. 94. Wada H, Wakita Y, Nakase T, et al: Hemostatic molecular
74. Genest J: C-reactive protein: risk factor, biomarker and/or markers before the onset of disseminated intravascular
therapeutic target? Can J Cardiol 26(suppl A):41A-44A, 2010. coagulation. Am J Hematol 60:273-278, 1999.
75. Glynn RJ, Koenig W, Nordestgaard BG, et al: Rosuvastatin 95. Francalance I, Comeglio P, Liotta AA, et al: D-dimer concen-
for primary prevention in older persons with elevated trations during normal pregnancy, as measured by ELISA.
C-reactive protein and low to average low-density lipopro- Thromb Res 80:89-92, 1995.
tein cholesterol levels: exploratory analysis of a randomized 96. Sawamura A, Hayakawa M, Gando S, et al: Disseminated
trial. Ann Intern Med 152:488-496, 2010. intravascular coagulation with a fibrinolytic phenotype at
76. Williams RH, Maggiore JA: Hyperhomocysteinemia patho- an early phase of trauma predicts mortality. Thromb Res
genesis, clinical significance, laboratory assessment, and 124:608-613, 2009.
treatment. Lab Med 30:468-474, 1999. 97. Moresco RN, Vargas LC, Voegeli CF, et al: D-dimer and
77. Homocysteinemia. In Hathaway WE, Goodnight SH, its relationship to fibrinogen/fibrin degradation products
editors: Disorders of hemostasis and thrombosis, ed 2, New (FDPs) in disorders associated with activation of coagula-
York, 2001, McGraw-Hill, pp 397-401. tion or fibrinolytic systems. J Clin Lab Anal 17:77-79,
78. Boers GHJ: Hyperhomocysteinemia as a risk factor for arte- 2003.
rial and venous disease: a review of evidence and relevance. 98. Boisclair MD, Lane DA, Wilde JT, et al: A comparative evalu-
Thromb Haemost 78:520-522, 1997. ation of assays for markers of activated coagulation and/or
79. Guba SC, Fonseca V, Fink LM: Hyperhomocysteinemia and fibrinolysis: thrombin-antithrombin complex, D-dimer and
thrombosis. Semin Thromb Hemost 25:291-309, 1999. fibrinogen/fibrin fragment E antigen. Br J Haematol 74:471-
80. Ciaccio M, Bellia C: Hyperhomocysteinemia and cardiovas- 479, 1990.
cular risk: effect of vitamin supplementation in risk reduc- 99. Favaloro EJ: Laboratory testing in disseminated intra
tion. Curr Clin Pharmacol 5:30-36, 2010. vascular coagulation. Semin Thromb Hemost 36:458-467,
81. Dangelo A, Selhub J: Homocysteine and thrombotic disease. 2010.
Blood 90:1-11, 1997. 100. Avvisati G, ten Cate JW, Buller HR, et al: Tranexamic acid
82. Cushman M, Alving BM: Risk factors for cardiovascular for control of haemorrhage in acute promyelocytic leuke-
disease and arterial thrombosis. In Kitchens CS, Alving MB, mia. Lancet 2:122-124, 1989.
Kessler CM, editors: Consultative hemostasis and thrombosis, 101. Speiser W, Mallek R, Koppensteiner R, et al: D-dimer
ed 2, Philadelphia, 2007, Saunders, pp 371-378. and TAT measurement in patients with deep venous
83. Meade TW, Mellows S, Brozovic M, et al: Haemostatic func- thrombosis: utility in diagnosis and judgment of anticoagu-
tion and ischaemic heart disease: principal results of the lant treatment effectiveness. Thromb Haemost 64:196-201,
Northwick Park Heart Study. Lancet 2:533-537, 1986. 1990.
CHAPTER 42 Thrombosis Risk Testing 693
102. Gorog DA: Prognostic value of plasma fibrinolysis activa- 107. Warkentin TE: Heparin-induced thrombocytopenia. In
tion markers in cardiovascular disease. J Am Coll Cardiol Colman RW, Marder VJ, Clowes AW, et al, editors: Hemostasis
15(55):2701-2709, 2010. and thrombosis: basic principles and clinical practice, ed 5,
103. Brace LD: Testing for heparin-induced thrombocytopenia by Philadelphia, 2006, Lippincott Williams & Wilkins,
platelet aggregometry. Clin Lab Sci 5:80-81, 1992. pp 1649-1662.
104. Vermylen J, Hoylaerts MF, Arnout J: Antibody-mediated 108. Warkentin TE, Sheppard JA: Testing for heparin-induced
thrombosis. Thromb Haemost 78:420-426, 1997. thrombocytopenia antibodies. Transfus Med Rev 20:259-
105. Crowther MA, Cook DJ, Albert M, et al: Canadian Critical 272, 2006.
Care Trials Group. The 4Ts scoring system for heparin- 109. Isenhaart CE, Brandt JT: Platelet aggregation studies for the
induced thrombocytopenia in medical-surgical intensive diagnosis of heparin-induced thrombocytopenia. Am J Clin
care unit patients. J Crit Care 25:287-293, 2010 Jun. Pathol 99:324-330, 1993.
106. Eby CS: Heparin induced thrombocytopenia. Clin Lab Sci
12:365-369, 1999.
43 Thrombocytopenia and
Thrombocytosis
Larry D. Brace

OUTLINE OBJECTIVES
Thrombocytopenia: After completion of this chapter, the reader will be able to:
Decrease in Circulating
Platelets 1. Define thrombocytopenia and thrombocytosis, and 7. Differentiate between neonatal isoimmune thrombocy-
Impaired or Decreased state their associated platelet counts. topenia and neonatal autoimmune thrombocytopenia.
Platelet Production 2. Compare and contrast the clinical symptoms of 8. Explain the laboratory findings and pathophysiology
Increased Platelet platelet disorders and clotting factor deficiencies. associated with thrombotic thrombocytopenic
Destruction 3. Explain the primary pathophysiologic processes of purpura and hemolytic uremic syndrome.
Abnormalities in thrombocytopenia. 9. Summarize the pathophysiology of thrombotic com-
Distribution or Dilution 4. Name and list the unique diagnostic features of at plications in heparin-induced thrombocytopenia and
Thrombocytosis: Increase least four disorders included in congenital hypoplasia describe the sequence of treatment options.
in Circulating Platelets of the bone marrow and describe their inheritance 10. Given clinical history and laboratory test results for
Reactive (Secondary)
patterns. patients with thrombocytopenia or thrombocytosis,
Thrombocytosis
5. Differentiate between acute and chronic immune suggest a diagnosis that is consistent with the infor-
Thrombocytosis
Associated with thrombocytopenia. mation provided.
Myeloproliferative 6. Describe the immunologic and nonimmunologic
Disorders mechanisms by which drugs may induce
thrombocytopenia.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

An 18-month-old African American girl sustained severe Patient Results Reference Range
burns over 40% to 50% of her body, including both lower Protein C antigen 78% 70-137%
extremities. Within 1 month, she underwent a below-knee Protein S antigen 120% 63-156%
amputation of the left lower extremity. Over the next several Antithrombin activity 111% 76-136%
years, she underwent skin-grafting surgeries, central venous
line placement, and other burn-related surgeries. During The patients results were normal on tests for the factor V
these procedures, the patient was exposed to heparinized Leiden and prothrombin G20210A mutations, and she was
saline irrigation. Four years after the burn injury, thrombosis found not to have antiphospholipid antibody syndrome. Her
was noted in the right femoral artery during a grafting surgery. platelet count had been decreasing steadily for 7 days before
Unfractionated heparin was used during the surgery. Sur- surgery but was still within the reference range.
geons were unable to save the leg, and an above-knee amputa- 1. Is the heparin used during the grafting surgeries significant
tion was necessary. At this time, hypercoagulability studies in this patients case?
were ordered. Results were as follows: 2. What test should be ordered next?

694
CHAPTER 43 Thrombocytopenia and Thrombocytosis 695

B
leeding disorders resulting from platelet abnormalities,
whether quantitative or qualitative, usually are mani- BOX 43-1 Classification of Thrombocytopenia
fested by bleeding into the skin or mucous membranes
or both (mucocutaneous bleeding). Common presenting Impaired or Decreased Production of Platelets
symptoms include petechiae, purpura, ecchymoses, epistaxis, Congenital
and gingival bleeding. Similar findings also are seen in vascular May-Hegglin anomaly
disorders, but vascular disorders (e.g., Ehlers-Danlos syn- Bernard-Soulier syndrome
drome, hereditary hemorrhagic telangiectasia) are relatively Fechtner syndrome
rare. In contrast, deep tissue bleeding, such as hematoma and Sebastian syndrome
hemarthrosis, is associated with clotting factor deficiencies. Epstein syndrome
Montreal platelet syndrome
Fanconi anemia
THROMBOCYTOPENIA: DECREASE Wiskott-Aldrich syndrome
IN CIRCULATING PLATELETS Thrombocytopenia with absent radii (TAR) syndrome
Although the reference range for the platelet count varies Congenital amegakaryocytic thrombocytopenia
among laboratories, it is generally considered to be approxi- Autosomal dominant and X-linked thrombocytopenia
mately 150,000 to 450,000/mcL (150,000 to 450,000/mm3 or Neonatal
150 to 450 109/L). Thrombocytopenia (platelet count of Acquired
fewer than 100,000/mcL) is the most common cause of clini- Viral
cally important bleeding. The primary pathophysiologic pro- Drug induced
cesses that result in thrombocytopenia are decreased platelet
production, accelerated platelet destruction, and abnormal Increased Platelet Destruction
platelet distribution (sequestration) (Box 43-1). Immune
Small-vessel bleeding in the skin attributed to thrombocy- Acute and chronic immune thrombocytopenic purpura
topenia is manifested by hemorrhages of different sizes (Figure Drug induced: immunologic
43-1). Petechiae are small pinpoint hemorrhages about 3mm Heparin induced thrombocytopenia
in diameter, purpura are about 1cm in diameter and generally Neonatal alloimmune (isoimmune neonatal) thrombocytopenia
round, and ecchymoses are 3cm or larger and usually irregular Neonatal autoimmune thrombocytopenia
in shape. Ecchymosis corresponds with the lay term bruise. Posttransfusion isoimmune thrombocytopenia
Clinical bleeding varies and often is not closely correlated with Secondary autoimmune thrombocytopenia
the platelet count. It is unusual for clinical bleeding to occur Nonimmune
when the platelet count is greater than 50,000/mcL, but the Thrombocytopenia in pregnancy and preeclampsia
risk of clinical bleeding increases progressively as the platelet Human immunodeficiency virus infection
count decreases from 50,000/mcL. Patients with platelet counts Hemolytic disease of the newborn
of 20,000/mcL or sometimes lower may have little or no bleed- Thrombotic thrombocytopenia purpura
ing symptoms. In general, patients with platelet counts of fewer Disseminated intravascular coagulation
than 10,000/mcL are considered to be at high risk for a serious Hemolytic uremic syndrome
hemorrhagic episode. Drug induced

Impaired or Decreased Platelet Production Abnormalities of Distribution or Dilution


Abnormalities in platelet production may be divided into two Splenic sequestration
categories: One type is associated with megakaryocyte hypo- Kasabach-Merritt syndrome
plasia in the bone marrow, and the other type is associated Hypothermia
with ineffective thrombopoiesis, as may be seen in disordered Loss of platelets: massive blood transfusions, extracorporeal circulation
proliferation of megakaryocytes. Data from Colvin BT: Thrombocytopenia, Clin Haematol 14:661-681, 1985;
and Thompson AR, Harker LA: Manual of hemostasis and thrombosis, ed 3,
Congenital Hypoplasia Philadelphia, 1983, FA Davis.
Lack of adequate bone marrow megakaryocytes (megakaryo-
cytic hypoplasia) is seen in a wide variety of congenital
disorders, including Fanconi anemia (pancytopenia), throm- May-Hegglin anomaly is a rare autosomal dominant disor-
bocytopenia with absent radius (TAR) syndrome, Wiskott- der; the exact frequency is unknown. Large platelets (20m in
Aldrich syndrome, Bernard-Soulier syndrome, May-Hegglin diameter) are present on the peripheral blood film, and Dhle
anomaly, and several other less common disorders. Although bodies are present in neutrophils (Figure 43-2) and occasion-
thrombocytopenia is a feature of Bernard-Soulier syndrome ally in monocytes. Other than the increase in size, platelet
and Wiskott-Aldrich syndrome, the primary abnormality in morphology is normal. Thrombocytopenia is present in about
these disorders is a qualitative defect, and these disorders are one third to one half of affected patients. Platelet function in
discussed in Chapter 44. response to platelet-activating agents is usually normal. In
696 PART VIII Hemostasis and Thrombosis

Epstein syndrome, large platelets are associated with deafness,


ocular problems, and glomerular nephritis.3
TAR syndrome is a rare autosomal recessive disorder char-
acterized by severe neonatal thrombocytopenia and congenital
absence or extreme hypoplasia of the radial bones of the fore-
arms with absent, short, or malformed ulnae and other ortho-
pedic abnormalities. The defects are presumed to occur as a
result of a specific type of fetal injury at about 8 weeks of gesta-
tion. TAR syndrome is one of the inherited impaired deoxyri-
bonucleic acid (DNA) repair disorders. Because exposure to
radiation causes cellular DNA damage, it is also considered to
be a radiation sensitivity syndrome.4 In addition, patients tend
to have cardiac lesions and a high incidence of transient leu-
kemoid reactions with elevated white blood cell (WBC) counts
(sometimes with counts above 100,000/mcL) in 90% of
patients.5 Platelet counts are usually 10,000 to 30,000/mcL in
Figure 43-1 Purpura and petechiae indicate systemic (mucocutaneous)
hemorrhage. (From Kitchens CS, Alving BM, Kessler CM: Consultative hemo- infancy. Interestingly, platelet counts usually increase over
stasis and thrombosis, Philadelphia, 2002, Saunders.) time, with normal levels often achieved within 1 year of birth.
Signaling through the thrombopoietin (TPO) receptor appears
to be abnormal, although the deficit has not been clearly
identified.
Fanconi anemia is also associated with thrombocytopenia,
although other abnormalities are extensive, including bony
abnormalities, abnormalities of visceral organs, and pancyto-
penia. Chapter 21 contains a more detailed description.

Congenital Amegakaryocytic Thrombocytopenia


Congenital amegakaryocytic thrombocytopenia is an autoso-
mal recessive disorder reflecting bone marrow failure.6 Affected
infants usually have platelet counts of fewer than 20,000/mcL
at birth, have petechiae and evidence of bleeding, and fre-
quently have physical anomalies. About half of the infants
develop aplastic anemia in the first year of life, and there are
reports of myelodysplasia and leukemia later in childhood.
Allogeneic stem cell transplantation is considered curative for
infants with clinically severe disease or aplasia.7 This disorder
is caused by mutations in the c-mlp gene resulting in complete
Figure 43-2 Dhle body in segmented neutrophil and giant platelets loss of thrombopoietin receptor function. This loss of function
associated with May-Hegglin anomaly (peripheral blood, 1000). (From results in reduced megakaryocyte progenitors and high throm-
Carr JH, Rodak BF: Clinical hematology atlas, ed 3, Philadelphia, 2009, bopoietin levels.8
Saunders.)
Autosomal Dominant and X-Linked
some patients, megakaryocytes are increased in number and Thrombocytopenia
have abnormal ultrastructure. Mutations in the MYH9 gene Autosomal dominant and X-linked thrombocytopenia are
that encodes for nonmuscle myosin heavy chain (a cytoskeletal well documented. In autosomal dominant thrombocytopenia,
protein in platelets) have been reported.1 This mutation may bleeding is usually mild, platelet function is normal, and the
be responsible for the abnormal size of platelets in this disor- megakaryocytes seem to be normal in number and morphol-
der. Most patients are asymptomatic unless severe thrombocy- ogy.9,10 X-linked thrombocytopenia can result from mutations
topenia is present, but bleeding times may be prolonged in in WASP (Wiskott-Aldrich syndrome protein) or mutations in
some patients in the absence of bleeding complications. the GATA1 gene.11-13 X-linked thrombocytopenias range from
Three other disorders involving mutations of the MYH9 mild thrombocytopenia and small platelets to macrothrombo-
gene have been reported: Sebastian syndrome, Fechtner syn- cytopenia with severe bleeding.
drome, and Epstein syndrome.2 Sebastian syndrome is inher-
ited as an autosomal dominant disorder characterized by large Neonatal Hypoplasia
platelets, thrombocytopenia, and granulocytic inclusions. Causes of neonatal megakaryocytic hypoplasia include
Similar abnormalities are observed in Fechtner syndrome and infection with Toxoplasma, rubella, cytomegalovirus (CMV),
are accompanied by deafness, cataracts, and nephritis. In and herpes (TORCH), human immunodeficiency virus (HIV)
CHAPTER 43 Thrombocytopenia and Thrombocytosis 697

infection, and in utero exposure to certain drugs, particularly hypertension, splenomegaly, and folic acid deficiency, should
chlorothiazide diuretics and the oral hypoglycemic tolbuta- be excluded. The platelet count usually returns to normal
mide and other agents. TORCH infections cause thrombocyto- within a few weeks of alcohol withdrawal, but thrombocyto-
penia with characteristically small platelets. CMV is the most penia may persist for longer periods. A transient rebound
common infectious agent causing congenital thrombocytope- thrombocytosis may develop when alcohol ingestion is
nia, with an overall incidence of 0.5% to 1% of all births,14 stopped.10
but only 10% to 15% of infected infants have symptomatic Interferon therapy commonly causes mild to moderate
disease,15 which suggests that the incidence of significant neo- thrombocytopenia, although under certain circumstances,
natal thrombocytopenia caused by CMV is about 1 in 1000 the thrombocytopenia can be severe and life-threatening.
infants. Although the mechanism of thrombocytopenia is not Interferon- and interferon- inhibit stem cell differentiation
well understood, reports suggest that CMV inhibits megakaryo- and proliferation in the bone marrow, but the mechanism of
cytes and their precursors, which results in impaired platelet action is unclear.28
production.16 About 1 in 1000 to 1 in 3000 infants are affected Thrombocytopenia presumably caused by megakaryocyte
by congenital toxoplasmosis. About 40% of such infants suppression also has been reported to follow the administra-
develop thrombocytopenia.17 Congenital rubella is now rare in tion of large doses of estrogen or estrogenic drugs such as
countries with organized immunization programs18,19; persis- diethylstilbestrol. Other drugs, such as certain antibacterial
tent thrombocytopenia is a prominent feature in infants with agents (e.g., chloramphenicol), tranquilizers, and anticonvul-
congenital rubella syndrome. Thrombocytopenia also is a sants, also have been associated with thrombocytopenia caused
feature of maternal transmission of HIV to the neonate and is by bone marrow suppression.29-31
a sign of intermediate to severe disease.20
Maternal ingestion of chlorothiazide diuretics or tolbuta- Ineffective Thrombopoiesis
mide can have a direct cytotoxic effect on the fetal marrow Thrombocytopenia is a usual feature of the megaloblastic
megakaryocytes. Thrombocytopenia may be severe, with plate- anemias (pernicious anemia, folic acid deficiency, and vitamin
let counts of 70,000/mcL and sometimes lower. Bone marrow B12 deficiency). Quantitative studies indicate that, as with
examination reveals a marked decrease or absence of mega- erythrocyte production in these disorders, platelet production
karyocytes. The thrombocytopenia develops gradually and is is ineffective. Although the bone marrow generally contains an
slow to regress when the drug is stopped. Recovery usually increase in the number of megakaryocytes, the total number of
occurs within a few weeks after birth.10,21,22 platelets released into the circulation is decreased. Thrombo-
cytopenia is caused by impaired DNA synthesis, and the bone
Acquired (Drug-Induced) Hypoplasia marrow may contain grossly abnormal megakaryocytes with
Drug-Induced Hypoplasia. A wide array of chemother- deformed, dumbbell-shaped nuclei, sometimes in large
apeutic agents used for the treatment of hematologic and numbers. Stained peripheral blood films reveal large platelets
nonhematologic malignancies suppress bone marrow mega- that may have a decreased survival time and may have abnor-
karyocyte production and the production of other hematopoi- mal function. Thrombocytopenia is usually mild, and there is
etic cells. Examples include the commonly used agents evidence of increased platelet destruction. Patients typically
methotrexate, busulfan, cytosine arabinoside, cyclophospha- respond within 1 to 2 weeks to vitamin replacement.22,32-34
mide, and cisplatin. The resulting thrombocytopenia may lead
to hemorrhage, and the platelet count should be monitored Miscellaneous Conditions
closely. Drug-induced thrombocytopenia is often the dose- Viruses are known to cause thrombocytopenia by acting on
limiting factor for many chemotherapeutic agents. Recombi- megakaryocytes or circulating platelets, either directly or in the
nant interleukin-11 has been approved for treatment of form of viral antigen-antibody complexes. Live measles vaccine
chemotherapy-induced thrombocytopenia, and thrombopoi- can cause degenerative vacuolization of megakaryocytes 6 to
etin may prove to be useful for this purpose.23-25 Zidovudine 8 days after vaccination. Some viruses interact readily with
(used for the treatment of HIV infection) is also known to platelets by means of specific platelet receptors. Other viruses
cause myelotoxicity and severe thrombocytopenia.26 associated with thrombocytopenia include CMV, varicella-
Several drugs specifically affect megakaryocytopoiesis zoster virus, rubella virus, Epstein-Barr virus (which causes
without significantly affecting other marrow elements. Anagre infectious mononucleosis), and the virus that causes Thai
lide is one such agent, although its mechanism of action is hemorrhagic fever.10
unknown. This characteristic has made anagrelide useful for Certain bacterial infections commonly are associated with
treating the thrombocytosis of patients with essential throm- the development of thrombocytopenia. This may be the result
bocythemia and other myeloproliferative disorders.27 of toxins of bacterial origin, direct interactions between bacte-
Ingestion of ethanol for long periods (months to years) ria and platelets in the circulation, or extensive damage to
may result in persistent severe thrombocytopenia. Although the endothelium, as in meningococcemia. Many cases of
the mechanism is unknown, studies indicate that alcohol thrombocytopenia in childhood result from infection. Purpura
can inhibit megakaryocytopoiesis in some individuals. Mild may occur in many infectious diseases in the absence of throm-
thrombocytopenia is a common finding in alcoholic patients, bocytopenia, presumably because of vascular damage (see
but other causes unrelated to ethanol use, such as portal Chapter 44).10,35
698 PART VIII Hemostasis and Thrombosis

A common cause of unexplained thrombocytopenia is infil- develop suddenly and there is no family history of hemor-
tration of the bone marrow by malignant cells with a progres- rhagic abnormalities or thrombocytopenia, the diagnosis of
sive decrease in marrow megakaryocytes as the abnormal ITP is almost certain. There is, at present, no specific test that
cells replace normal marrow elements. Inhibitors of thrombo- is diagnostic of acute or chronic ITP.
poiesis may be produced by these abnormal cells and may help In mild cases of acute ITP, patients may have only scattered
to account for the thrombocytopenia associated with condi- petechiae. In most cases of acute ITP, however, patients develop
tions such as myeloma, lymphoma, metastatic cancer, and fairly extensive petechiae and some ecchymoses and may have
myelofibrosis.22,32,36 hematuria or epistaxis or both. About 3% to 4% of acute ITP
cases are considered severe, and typically generalized purpura
Increased Platelet Destruction is present, often accompanied by gastrointestinal bleeding,
Thrombocytopenia as a result of increased platelet destruction hematuria, mucous membrane bleeding, and retinal hemor-
can be separated into two categories: increased platelet destruc- rhage. Of patients with severe disease, 25% to 50% are consid-
tion caused by immunologic responses and increased destruc- ered to be at risk for intracranial hemorrhage, which is the
tion caused by mechanical damage or consumption or both. primary complication that contributes to the overall 1% to 2%
Regardless of the process, increased production is required to mortality rate for patients with acute ITP.38 Most patients with
maintain a normal platelet count, and the patient becomes life-threatening hemorrhage have a platelet count of fewer than
thrombocytopenic only when production capacity is no longer 4000/mcL.39 Hemorrhage is rarely experienced by patients
adequate to compensate for the increased rate of destruction. whose platelet count exceeds 10,000/mcL.
Most patients with acute ITP recover with or without treat-
Immune Mechanisms of Platelet Destruction ment in about 3 weeks, although for some, recovery may take
Immune (Idiopathic) Thrombocytopenic Purpura: 6 months. In a few children, recurrent episodes of acute ITP
Acute and Chronic. The term idiopathic thrombocytopenic are occasionally seen after complete recovery from the first
purpura (ITP) was used previously to describe cases of throm- episode.40 Most patients with acute ITP have relatively mild
bocytopenia arising without apparent cause or underlying symptoms, and no treatment is needed. The most severe cases
disease state. Although the acronym for the disorder remains may need to be treated, however, and intravenous immuno-
the same, the word idiopathic has been replaced by immune globulin (IVIG), platelet transfusions, and splenectomy (or
because of the realization that acute and chronic ITP are immu- some combination of these) seem to offer the most immediate
nologically mediated. benefit.37,38
Acute ITP is primarily a disorder of children, although a Chronic ITP can be found in patients of any age, although
similar condition is seen occasionally in adults. The disorder most cases occur in patients between the ages of 20 and 50
is characterized by the abrupt onset of bruising, petechiae, and years. Females with this disorder outnumber males 2:1 to 3:1,
sometimes mucosal bleeding (e.g., epistaxis) in a previously with the highest incidence in women between 20 and 40 years
healthy child. The primary hematologic feature is throm of age. The incidence of chronic ITP ranges from 3.2 to 6.6
bocytopenia, which frequently occurs 1 to 3 weeks after an cases per 100,000 per year.41 Chronic ITP usually begins insidi-
infection. ously, with platelet counts that are variably decreased and
The infection is most often a nonspecific upper respiratory sometimes normal for periods of time. Presenting symptoms
tract or gastrointestinal tract viral infection, but acute ITP also are those of mucocutaneous bleeding, with menorrhagia,
may occur after rubella, rubeola, chickenpox, or other viral recurrent epistaxis, and easy bruising (ecchymoses) being most
illnesses and may follow live virus vaccination.37 The incidence common.
of acute ITP is estimated to be 4 in 100,000 children, with a Platelet destruction in chronic ITP is the result of an immu-
peak frequency in children between 2 and 5 years of age. There nologic process. The offending antibodies attach to platelets,
is no sex predilection. In about 10% to 15% of the children and as a result, the antibody-labeled platelets are removed
initially thought to have acute ITP, the thrombocytopenia per- from the circulation by reticuloendothelial cells, primarily in
sists for 6 months or longer, and these children are reclassified the spleen. Autoantibodies that recognize platelet surface
as having chronic ITP.38 The observation that acute ITP often glycoproteins such as glycoprotein IIb (GP IIb) and GP IIIa
follows a viral illness suggests that some children produce (IIb/3), GP Ia/IIa, and others can be demonstrated in 50%
antibodies and immune complexes against viral antigens and to 60% of ITP patients.42,43 Because megakaryocytes also express
that platelet destruction may result from the binding of these GP IIb/IIIa and GP Ib/IX on their membranes, these cells are
antibodies or immune complexes to the platelet surface. obvious targets of the antibodies. Platelet turnover studies have
The diagnosis of acute ITP in a child with severe thrombo- shown impaired platelet production in ITP. Overall, the life
cytopenia almost always can be made without bone marrow span of the platelet is shortened from the normal 7 to 10 days
examination. If the child has recent onset of bleeding signs and to a few hours, and the rapidity with which platelets are
symptoms, otherwise normal results on complete blood count removed from the circulation correlates with the degree of
(for all red blood cell [RBC] and WBC parameters and cell thrombocytopenia. If plasma from a patient with ITP is
morphology), and normal findings on physical examination infused into the circulation of a normal recipient, the recipient
(except for signs of bleeding), there is a high likelihood that develops thrombocytopenia. The thrombocytopenia-producing
the child has ITP. In addition, if the bleeding symptoms factor in the plasma of the ITP patient is an immunoglobulin
CHAPTER 43 Thrombocytopenia and Thrombocytosis 699

chronic ITP consists principally of prednisone. About 70% to


90% of patients respond to this therapy with an increase
in platelet count and a decrease in hemorrhagic episodes.
Although reported response rates vary widely, about 50% of
patients have a long-term beneficial effect from corticosteroid
treatment.46 If the response to corticosteroids is inadequate or
no response is seen, steroid therapy can be supplemented
with IVIG or, in some cases, anti-D immunoglobulin.47,48 For
patients in whom prednisone is ineffective, intravenous ritux-
imab should be tried. Responses to rituximab are usually seen
within 3 to 4 weeks. In some patients splenectomy may become
necessary. Splenectomy eliminates the primary site of platelet
removal and destruction, but it also removes an organ contain-
ing autoantibody-producing lymphocytes. Splenectomy is an
effective treatment for adult chronic ITP, with 88% of patients
Figure 43-3 Typical peripheral blood cell morphology in immune throm- showing improvement and 66% having a complete and lasting
bocytopenic purpura. Note scarce platelets and increased platelet size but
response.49 Vaccination with pneumococcal, meningococcal,
normal red blood cell and leukocyte morphology (peripheral blood, 500).
and H influenza vaccines should be performed 2 weeks prior
to surgery. In the most severe refractory cases, immunosuppres-
G (IgG) antibody that can be removed from serum by adsorp- sive (chemotherapeutic) agents such as azathioprine given
tion with normal human platelets. In addition, cytotoxic T alone or with steroids may be necessary. In such patients,
cellmediated lysis of platelets has been shown in vitro using platelet transfusions may be of transient benefit in treating
CD3+CD8+ lymphocytes from patients with active chronic severe hemorrhagic episodes but should not be given rou-
ITP, although the in vivo significance of this mechanism is tinely.45 IVIG given alone or just before platelet transfusion
not known.44 also may be beneficial.37,45
The only abnormalities in the peripheral blood of patients Chronic ITP occurring in association with HIV infection,
with ITP are related to platelets. In most cases, platelets number with hemophilia, or with pregnancy presents special problems
between 30,000/mcL and 80,000/mcL. Patients with ITP in diagnosis and therapy. Unexplained thrombocytopenia in
undergo periods of remission and exacerbation, however, and otherwise healthy members of high-risk populations may be
their platelet counts may range from near-normal to fewer than an early manifestation of acquired immune deficiency syn-
20,000/mcL during these periods (Figure 43-3). Morphologi- drome (AIDS).36,45
cally, platelets appear normal, although larger in diameter than Differentiation of acute versus chronic idiopathic
usual. This is reflected in an increased mean platelet volume as thrombocytopenic purpura. The differences between acute
measured by electronic cell counters. The marrow typically is and chronic ITP are summarized in Table 43-1. Acute ITP
characterized by megakaryocytic hyperplasia. Megakaryocytes occurs most frequently in children 2 to 9 years of age and in
are increased in size, and young forms with a single nucleus, young adults, whereas chronic ITP occurs in patients of all ages,
smooth contour, and diminished cytoplasm are commonly although most frequently in adults aged 20 to 50 years and
seen. In the absence of bleeding, infection, or other underlying more commonly in women. Of patients with acute ITP, 60%
disorder, erythrocyte and leukocyte precursors are normal in to 80% have a history of infection, usually viral (rubella,
number and morphology. Coagulation tests showing abnor- rubeola, chickenpox, and nonspecific respiratory tract infec-
mal results include tests dependent on normal platelet func- tion), occurring 2 to 21 days before ITP onset. Acute ITP also
tion, such as bleeding time and clot retraction time. Although may occur after immunization with live vaccine for measles,
platelet-associated IgG levels are increased in most patients,10,21,45 chickenpox, mumps, and smallpox.
it has not been shown conclusively that any method of testing Acute ITP usually is self-limited, and spontaneous remis-
for platelet antibodies is sensitive or specific for ITP. sions occur in 80% to 90% of patients, although the duration
The initial treatment of chronic ITP depends on the urgency of the illness may range from days to months. In chronic ITP,
for increasing the platelet count. In ITP patients with platelet there is typically a fluctuating clinical course, with episodes of
counts greater than 30,000/mcL who receive no treatment, the bleeding that last a few days or weeks, but spontaneous remis-
expected mortality rate is equal to that of the general popula- sions are uncommon and usually incomplete.45
tion. Unless there are additional risk factors, ITP patients with Symptoms of acute ITP vary, but petechial hemorrhages,
platelet counts greater than 30,000/mcL should not be treated. purpura, and often bleeding from the gums and gastrointesti-
If additional risk factors are present, such as old age, coagula- nal or urinary tract typically begin suddenly, sometimes over a
tion defects, recent surgery, trauma, or uncontrolled hyperten- few hours. Hemorrhagic bullae in the oral mucosa are often
sion, the platelet count should be kept at 50,000/mcL or higher prominent in patients with severe thrombocytopenia of acute
depending on the clinical situation. In patients in whom the onset. Usually the severity of bleeding is correlated with the
need is considered urgent, IVIG remains the treatment of degree of thrombocytopenia.45 In contrast, presenting symp-
choice. For most patients, however, the initial treatment of toms of chronic ITP begin with a few scattered petechiae or
700 PART VIII Hemostasis and Thrombosis

TABLE 43-1 Clinical Picture of Acute and Chronic


BOX 43-2 Common Drugs Causing Immune
Immune Thrombocytopenic Purpura
Thrombocytopenia
Characteristic Acute Chronic Analgesics
Age at onset 2-6yr 20-50yr Salicylates
Sex predilection None Female over male, 3:1 Acetaminophen
Prior infection Common Unusual Phenylbutazone
Onset of bleeding Sudden Gradual Antibiotics
Platelet count <20,000/mcL 30,000-80,000/mcL Cephalothin
Duration 2-6wk Months to years Penicillin
Spontaneous remission 90% of patients Uncommon Streptomycin
Seasonal pattern Higher incidence in None Aminosalicylic acid
winter and spring Rifampin
Therapy Novobiocin
Steroids 70% response rate 30% response rate Various sulfa drugs (chlorthalidone, furosemide)
Splenectomy Rarely required <45yr, 90% response Alkaloids
rate; >45yr, 40% Quinidine
response rate Quinine
Sedatives, anticonvulsants
Data from Triplett DA, editor: Platelet function: laboratory evaluation and clinical Methoin
application, Chicago, 1978, American Society of Clinical Pathologists; Quick AJ: Hem- Troxidone
orrhagic diseases and thrombosis, ed 2, Philadelphia, 1966, Lea & Febiger; and Bussel
J, Cines D: Immune thrombocytopenia, neonatal alloimmune thrombocytopenia, and Chlorpromazine
post-transfusion purpura. In Hoffman R, Benz EJ Jr, Shattil SJ, et al, editors: Hematol- Diphenylhydantoin
ogy: basic principles and practice, ed 3, New York, 2000, Churchill Livingstone, Meprobamate
pp 2096-2114.
Phenobarbital
Carbamazepine
other minor bleeding manifestations. Occasionally, a bruising Oral hypoglycemics
tendency, menorrhagia, or recurrent epistaxis is present for Chlorpropamide
months or years before diagnosis. Platelet counts range from Tolbutamide
5000/mcL to 75,000/mcL and are generally higher than those Heavy metals
in acute ITP. Giant platelets are commonly seen. Platelet- Gold
associated immunoglobulin levels are elevated in most patients, Mercury
but the test lacks sensitivity and specificity.45 Bismuth
Treatment also varies for acute and chronic ITP. In chronic Organic arsenicals
ITP, initial therapy often consists of glucocorticoids (cortico- Miscellaneous
steroids), which interfere with splenic and hepatic macro- Chloroquine
phages to increase platelet survival time. If the ITP does not Chlorothiazide
respond to corticosteroids or the patient cannot tolerate them Insecticides
because of the resultant immunosuppression and toxicity, sple- Data from Triplett DA, editor: Platelet function: laboratory evaluation and clinical
nectomy may be necessary. In acute ITP, treatment for all but application, Chicago, 1978, American Society of Clinical Pathologists; and Quick
the most severely thrombocytopenic and hemorrhagic patients AJ: Hemorrhagic diseases and thrombosis, ed 2, Philadelphia, 1966, Lea &
is contraindicated. When treatment is necessary, a good Febiger.
response to IVIG or corticosteroids or both usually can be
obtained, and splenectomy is rarely required.37,38 antibody production has begun, the platelet count falls rapidly
and often may be fewer than 10,000/mcL. Patients may have
Immunologic Drug-Induced Thrombocytopenia. As abrupt onset of bleeding symptoms. If this type of drug-
can be seen from Box 43-2, many drugs can induce acute induced thrombocytopenia develops in a pregnant woman,
thrombocytopenia. Drug-induced immune-mediated throm- both she and her fetus may be affected. Quinine previously was
bocytopenia can be divided into several types based on the used to facilitate labor but is no longer used for this purpose.
interaction of the antibody with the drug and platelets. Mecha- The initial studies of quinidine-induced thrombocytopenia
nisms of drug-antibody binding are shown in Figure 43-4. suggested that the drug first combines with the antibody and
One type is typified by quinidine- and quinine-induced that the antigen-antibody (immune) complex then attaches to
thrombocytopenia and has been recognized for more than 100 the platelet in an essentially nonspecific manner (the inno-
years. The antibody induced by drugs of this type interacts with cent bystander hypothesis). It now seems clear, however, that
platelets only in the presence of the drug. Many drugs can the antibodies responsible for drug-induced thrombocytope-
induce such antibodies, but quinine, quinidine, and sulfon- nia bind to the platelets by their Fab regions, rather than by
amide derivatives do so more often than other drugs. When attaching nonspecifically as immune complexes. The innocent
CHAPTER 43 Thrombocytopenia and Thrombocytosis 701

bystander/immune complex explanation for this type of drug- specific target on the platelet membrane without covalently
induced thrombocytopenia should be abandoned. The Fab linking to the target or the antibody remains to be determined,
portion of the antibody binds to a platelet membrane constitu- however. Because the Fc portion of the immunoglobulin is not
ent, usually the GP Ib/IX complex or the GP IIb/IIIa complex, involved in binding to platelets, it is still available to the Fc
only in the presence of drug.50,51 The mechanism by which the receptors on phagocytic cells. This situation may contribute to
drug promotes binding of a drug-dependent antibody to a the rapid onset and relatively severe nature of the thrombocy-
topenia. Most drug-induced platelet antibodies are of the IgG
class, but in rare instances, IgM antibodies are involved.45
A second mechanism of drug-induced thrombocytopenia is
Macrophage induction of hapten-dependent antibodies. Some drug mole-
Fc receptor
cules are too small by themselves to trigger an immune
response, but they may act as a hapten and combine with a
larger carrier molecule (usually a plasma protein or protein
constituent of the platelet membrane) to form a complex that
can act as a complete antigen.45 Penicillin and penicillin deriva-
tives are the primary offending agents causing drug-induced
Fc receptor
thrombocytopenia by this mechanism. Drug-induced throm-
bocytopenia of this type is often severe. The initial platelet
count may be fewer than 10,000/mcL and sometimes fewer
than 1000/mcL. The number of bone marrow megakaryocytes
Platelet is usually normal to elevated.45 Bleeding is often severe and
rapid in onset, and hemorrhagic bullae in the mouth may be
prominent.
Drug-induced autoantibodies represent a third mechanism
of drug-induced thrombocytopenia. In this case, the drugs
stimulate the formation of an autoantibody that binds to a
specific platelet membrane glycoprotein with no requirement
Site on Fc segment of IgG binding to Fc receptors
for the presence of free drug. Gold salts and procainamide are
Structural antigenic determinant of the platelet membranes two examples of such drugs. Levodopa also may cause throm-
bocytopenia in the same way. The precise mechanism by which
Antigen of plasma immune complex
these drugs induce autoantibodies against platelets is not
Drug known with certainty.
Heparin-induced thrombocytopenia (HIT) is a good
Drug + platelet surface binding site
example of another type of drug-induced thrombocytopenia.
Figure 43-4 Immunoglobulin binds a platelet membrane antigen or Heparin binds to platelet factor 4 (PF4), a heparin-neutralizing
antigen and drug combination. Macrophage Fc receptors bind the Fc portion protein made and released by platelets (Figure 43-5). Binding
of the immunoglobulin. This may result in platelet removal and thrombocy- of heparin by plasma PF4 or platelet membraneexpressed PF4
topenia. IgG, Immunoglobulin G. (From Rapaport SI: Introduction to hematol- causes a conformational change in PF4, resulting in the expo-
ogy, ed 2, Philadelphia, 1987, JB Lippincott, p 489.) sure of neoepitopes. Exposure of these neoepitopes (new

Circulating PF4- Circulating PF4-


heparin complexes heparin-Ab complexes

PF4 neoepitope + Immune Ab to PF4


and/or exposure response neoepitopes and/or

Platelet surface PF4- Platelet surface PF4-


heparin complexes heparin-Ab complexes

Ab Fc attaches to
Platelet agglutination, aggregation, and activation
platelet Fc receptor
occludes vessel with platelet thrombus

Figure 43-5 Heparin-induced thrombocytopenia with thrombosis. An antibody (Ab) binds the heparinplatelet factor 4 (PF4) complex in plasma or on the
platelet surface. The Fc portion binds platelet Fc receptors and activates the platelet. The activated platelets aggregate to form platelet thrombi in the arterial
circulation.
702 PART VIII Hemostasis and Thrombosis

antigens) stimulates the immune system of some individuals, A common benign form of HIT that occurs on heparin
which leads to the production of an antibody to one of the administration is type I (nonimmune mediated) HIT. It is
neoepitopes. In HIT, heparin and PF4 form a complex on the important to distinguish benign type I from type II HIT. Type
platelet surface or circulating free complexes to which the anti- I is associated with a rapid decrease in the platelet count after
body binds. The Fab portion of the immunoglobulin molecule administration of heparin, but the thrombocytopenia is mild
binds to an exposed neoepitope in the PF4 molecule; this (the platelet count rarely decreases to fewer than 100,000/mcL)
leaves the Fc portion of the IgG free to bind with the platelet and transient, and the platelet count returns rapidly to the
FcR receptor, which causes platelet activation.52,53 Because the preheparin level even if heparin therapy is continued. Careful
Fc portions of the IgG molecules bind to platelet FcR recep- attention should be paid to the platelet count and other signs
tors, they are not available to the Fc receptors of the cells of of HIT so that this form of thrombocytopenia is not confused
the reticuloendothelial system. This may explain the less severe with the clinically significant type II HIT. Although the mecha-
decline in platelet count in this thrombocytopenia. That does nism of type I HIT has not been completely described, it may
not mean, however, that the consequences are less serious. The be related to the well-documented proaggregatory effects of
opposite may be true. Because platelets are activated by occu- heparin.54,57 Because activated or aggregated platelets are
pancy of their FcR receptors, in vivo platelet aggregation with cleared from the circulation, these effects may explain the mild
thrombosis is possible. HIT sometimes is referred to as heparin- decrease in the platelet count that occurs during the first few
induced thrombocytopenia and thrombosis. Heparin binding to days of heparin administration. This has not been clearly docu-
PF4 is required to expose the neoepitope to which the antibody mented, however.
binds. The treatment for HIT is to discontinue heparin admin- The binding of heparin and related compounds depends on
istration and replace it with another suitable anticoagulant. polysaccharide chain length, composition, and degree of sul-
Low-molecular-weight heparin should not be used as a heparin fation. Short-chain heparin polysaccharides (low-molecular-
replacement for this purpose, because the antibody cross-reacts weight heparin) have lower affinity to PF4 and are less prone
with low-molecular-weight heparin and PF4 to result in plate- to cause type II HIT. Pentasaccharide and its synthetic deriva-
let activation and aggregation.54 tives (e.g., fondaparinux or idraparinux) do not seem to bind
HIT is a relatively common side effect of unfractionated PF4 and are unlikely to cause type II HIT.
heparin administration, with about 1% to 5% of patients The detection of clinically significant HIT by laboratory
developing this complication. Despite the thrombocytopenia, testing is problematic, because all tests lack sensitivity. Three
patients with HIT usually are not at significant risk of bleeding, methods are commonly used, but all depend on the presence
because the platelet count typically does not fall below 40,000/ of free heparin-induced antiplatelet antibodies in the patients
mcL. Ten percent to 30% of patients with HIT develop throm- serum or plasma in sufficient quantity to cause a positive test
botic complications, however. In patients who develop HIT, result. HIT can be detected by a platelet aggregation tech-
heparin therapy should be stopped as soon as the diagnosis is nique.58 In this method, serum from the patient is added to
made, because continued heparin therapy can lead to signifi- platelet-rich plasma from normal donors, heparin is added to
cant morbidity and mortality, including gangrene of the the mixture, and platelet aggregation is typically monitored for
extremities, amputation, and death. After discontinuation of 20 minutes. The specificity of the method is excellent (near
heparin, the platelet count begins to increase and should return 100%), but the sensitivity is quite low (about 50%). The sen-
to normal within a few days.55 sitivity of the test can be improved, but this requires the use of
Because the immune system is involved in the development several heparin concentrations and of platelet-rich plasma
of HIT, the clinical signs of HIT typically are not seen until 7 from two or more blood donors, preferably of the same ABO
to 14 days after the initiation of heparin therapy (the time blood type as the patient. In addition, the individuals donating
necessary to mount an immune response on first exposure to blood for platelet-rich plasma must not have taken aspirin for
an antigen). If the patient has been exposed to heparin previ- 10 to 14 days before platelet donation, because platelets from
ously, however, symptoms of HIT may be seen in 1 to 3 days. donors who have ingested aspirin produce a false-negative
Because the platelet count may fall sharply in 1 day, it is recom- result for HIT.54 In the authors experience, patients who
mended that platelet counts be measured daily in patients develop HIT while receiving aspirin therapy rarely develop
receiving unfractionated heparin therapy. One other sign of thrombotic complications, and the drop in their platelet counts
impending HIT in some patients is the development of heparin is less precipitous, gradually decreasing over the course of
resistance. This is the clinical situation in which a patient who several days (unpublished observations). Given the efficacy of
had experienced adequate anticoagulation at a certain heparin aspirin therapy in primary and secondary prevention of myo-
dosage suddenly requires increasing amounts of heparin to cardial infarction and thrombotic stroke, it is increasingly dif-
maintain the same level of anticoagulation. This situation can ficult to find a sufficient pool of suitable (and willing) blood
result from in vivo activation of platelets and release of PF4 donors. Although the procedure is time consuming, reasonable
and -thromboglobulin from platelet -granules. Both of these sensitivity can be obtained with sufficient attention to the
substances neutralize heparin, which leads to a normalization details of the technique.27
of results on the partial thromboplastin time test that is used Dense granules of platelets contain serotonin, and platelets
to monitor heparin therapy. Heparin resistance often is seen have an active mechanism for rapid uptake and storage of
before the development of thrombocytopenia.56 serotonin in dense granules. This property of platelets is used
CHAPTER 43 Thrombocytopenia and Thrombocytosis 703

No 3H-serotonin
release
Same ABO blood group ()
as patient preferred

3H-serotonin
+ Therapeutic heparin
concentration release

3H-labeled +Patient
Donor PRP+ 3H-serotonin No 3H-serotonin HIT
donor platelets serum
release

+ Supertherapeutic
heparin concentration
3H-serotonin
release
()

Figure 43-6 The serotonin release assay for heparin-induced thrombocytopenia (HIT). Donor platelets in platelet-rich plasma (PRP) are labeled with tritiated
(3H) serotonin, washed, and suspended in patient plasma. Heparin in therapeutic and saturating doses is added to two aliquots. Release of radioactive serotonin
in the therapeutic aliquot in combination with no release in the supratherapeutic system indicates HIT.

PF4-coated PF4-heparin PF4-heparin-Ab


microtiter Heparin complexes Patient complexes if Ab
plate wells on surfaces serum to PF4 is present

Enzyme-labeled
antihuman Ab
No color
(negative)

PF4-heparin-Ab-enzyme-labeled Substrate with


antihuman Ab complexes chromophore
Color development
if Ab to PF4 is present (positive)

Figure 43-7 Enzyme immunoassay for heparin-induced thrombocytopenia. The solid-phase target antigen is a complex of platelet factor 4 (PF4) and
heparin or a heparin surrogate. Antiheparin-PF4 in patient serum binds the antigen and is bound by enzyme-labeled antihuman antibody (Ab), a sandwich
assay. The enzyme catalyzes the release of a chromophore from its substrate.

in another test for HIT in which washed normal platelets from requirement for radiolabeled serotonin. Most clinical labora-
a donor are incubated with radioactive serotonin (Figure tories no longer use isotopic techniques. As a result, this test is
43-6).59 Radioactive serotonin is taken up rapidly and stored performed only in a few specialized centers. In addition, it has
in the dense granules of the donor platelets, which are washed some of the same methodologic drawbacks as the platelet
and resuspended. In the presence of heparin-dependent anti- aggregation technique, in particular the need for platelets from
platelet antibody and heparin, the donor platelets become drug-free donors. Nonetheless, when properly performed, this
activated and release the contents of their dense granules when technique has similar specificity and superior sensitivity com-
the concentration of heparin in the test suspension is near the pared with the platelet aggregation method.
therapeutic range. The reappearance of radioactive serotonin More recently, an enzyme-linked immunosorbent assay has
in the plasma indicates the presence of a heparin-dependent been developed based on the knowledge that the antigenic
antiplatelet antibody (i.e., HIT). Under these same conditions, target of the heparin-dependent antiplatelet antibody is a PF4-
supratherapeutic concentrations of heparin do not activate heparin complex (Figure 43-7). In this assay, PF4 (or a related
platelets, however, and if platelets release the contents of dense compound) is coated to the surfaces of microplate wells.
granules at both therapeutic and supratherapeutic concentra- Heparin and the serum or plasma from the patient suspected
tions of heparin, the test result is not positive for HIT. A similar to have HIT are added to wells of the microtiter plate. If the
phenomenon is observed in the test that uses platelet antibody is present, it adheres to PF4 in the presence of heparin
aggregometry.54,60 This method is commonly called the sero- or heparin-like compounds. The plate wells are washed, and
tonin release assay and, before the development of enzyme- enzyme-labeled monoclonal antibodies against human IgG
linked immunosorbent assays for HIT, was considered the and IgM are added. After an appropriate incubation period, the
gold standard for detection of HIT. Its major drawback is the plate is washed, and a chromogenic substrate for the enzyme
704 PART VIII Hemostasis and Thrombosis

is added. Color development in the assay well indicates the thrombocytopenias. Laboratory testing to identify the specific
presence of the heparin-dependent antiplatelet antibody in drug involved is usually beyond the capabilities of most labo-
the patient plasma.61 This assay has greater sensitivity than the ratories. This type of testing is performed by many reference
platelet aggregation method and similar sensitivity to the sero- laboratories, however.
tonin release assay, but it has lower specificity than the sero-
tonin release assay or the platelet aggregation method. Unlike Neonatal Alloimmune Thrombocytopenia. Neonatal
the functional HIT assays (serotonin release assay and platelet alloimmune thrombocytopenia (NAIT) develops when the
aggregation) the enzyme-linked immunosorbent assay (ELISA) mother lacks a platelet-specific antigen (usually human platelet
can detect antiPF4-heparin antibodies that are not pathologic; antigen 1a, or HPA-1a [P1A1]) that the fetus has inherited from
that is, the test result is positive but the patient does not have the father. Fetal platelet antigens may pass from the fetal to the
clinical HIT. The first ELISA assays detected both IgG and IgM maternal circulation as early as the fourteenth week of gesta-
antibodies to heparinPF4 complexes. It appears that IgM anti- tion.62 If the mother is exposed to a fetal antigen she lacks, she
bodies are not as clinically relevant and may be the cause of may make antibodies to that fetal antigen. These antibodies
false-positives tests for HIT. More recently, ELISA kits that cross the placenta, attach to the antigen-bearing fetal platelets,
detect only IgG antibodies have been developed and seem to and result in thrombocytopenia in the fetus. In this regard, the
have greater specificity (less clinically false-positive test results). pathophysiology of NAIT is the same as that of hemolytic
The enzyme-linked immunosorbent assay method is consider- disease of the newborn. The most frequent cause of NAIT in
ably less labor intensive, does not require blood from healthy whites is the HPA-1a antigen expressed on GP IIIa of the
drug-free donors, and can be performed in most laboratories. surface membrane GP IIb/IIIa complex, followed by HPA-5b
For patients who develop type II HIT, it is essential that (Bra). The antigen HPA-3a (Baka) is present on GP IIb and is
heparin therapy be withdrawn immediately. It is not prudent, an important cause of neonatal thrombocytopenia in Asians.
however, to discontinue administration of an anticoagulant/ Platelet antigen HPA-4 (Penn and Yuk) accounts for the disor-
antithrombotic without substituting a suitable alternative. It is der in a few affected neonates. Clinically significant thrombo-
clear from the literature that under these circumstances, with- cytopenia develops in an estimated 1 in 1000 to 2000
drawal of heparin treatment without replacement anticoagu- newborns.63 With the first pregnancy, about 50% of neonates
lant therapy results in an unacceptably high rate of thrombotic born to mothers lacking a specific platelet antigen are affected,
events. In the recent past, good alternatives for heparin were whereas with subsequent pregnancies the risk is 75% to 97%.63
not available. Today, several alternative agents are suitable sub- The incidence of intracranial hemorrhage or death or both in
stitutes (although considerably more expensive), including affected offspring is about 25%, and about half of the intracra-
direct thrombin inhibitors such as hirudin and its analogues, nial hemorrhages occur in utero in the second trimester.
argatroban and related synthetic direct thrombin inhibitors, Affected infants may appear normal at birth but soon manifest
and fondaparinux (Arixtra) and related synthetic pentasaccha- scattered petechiae and purpuric hemorrhages. In many infants
rides (e.g., idraparinux). Fondaparinux is identical in chemical with NAIT, serious hemorrhage does not develop, however,
structure to the antithrombin binding sequence in heparin and and the infants recover over a 1- to 2-week period as the level
heparin-derived agents such as low-molecular-weight heparins, of passively transferred antibody decreases.10,38 In symptomatic
while idraparnux is a chemically modified analog of the same cases, platelet levels are usually below 30,000/mcL and may
structure. diminish even further in the first few hours after birth.
Treatment for any drug-induced thrombocytopenia is first The diagnosis of NAIT is one of exclusion of other causes
to identify the offending drug, immediately discontinue its use, of neonatal thrombocytopenia, including maternal ITP and
and substitute another suitable therapeutic agent. This is often maternal ingestion of drugs known to be associated with drug-
difficult to accomplish. Many patients are taking multiple induced thrombocytopenia. The presence of thrombocytope-
drugs, and it is not always easy to determine which of the drugs nia in a neonate with a HPA-1a-negative mother or a history
is at fault. Under these conditions, identifying the causative of the disorder in a sibling is strong presumptive evidence in
agent may be a trial-and-error procedure in which the most favor of the diagnosis. Confirmation should include platelet
likely drugs are eliminated one at a time. In addition, even if typing of both parents and testing for evidence of a maternal
the patient is taking only one agent, there may not be a suitable antibody directed at paternal platelets.37
replacement, or a prolonged period may be required for the In situations in which suspicion of NAIT is high or there is
alternative drug to become effective. Drugs usually are cleared a history of NAIT in a first pregnancy, it may be necessary to
from the circulation rapidly, but dissociation of drug-antibody test or treat the fetus to prevent intracranial hemorrhage in
complexes may require longer periods, perhaps 1 to 2 weeks.10 utero. Fetal genotypes now can be determined at 10 to 18
In some cases, such as those caused by gold salts, thrombocy- weeks of gestation using polymerase chain reaction methods
topenia may persist for months. Platelet transfusions may be on cells obtained by chorionic villus sampling or amniocente-
necessary for patients with life-threatening bleeds. Although it sis.64 Periumbilical sampling to determine the fetal platelet
is true that the transfused platelets are destroyed rapidly, they count can be performed at about 20 weeks of gestation. When
may function to halt bleeding effectively before they are the fetus is thrombocytopenic, weekly maternal infusions of
destroyed. In addition, high-dose IVIG may be used and is IVIG have been shown to be effective in increasing the fetal
generally an effective treatment for most drug-induced immune platelet count in most cases.65 In cases in which the fetal
CHAPTER 43 Thrombocytopenia and Thrombocytosis 705
platelet count does not increase with IVIG therapy, washed washed RBCs. PTP is manifested by the rapid onset of severe
maternal platelets have been infused into the fetus with good thrombocytopenia and moderate to severe hemorrhage that
results.66 Treatment of the mother with high-dose corticoste- may be life-threatening. The recipients plasma is found to
roids (to decrease maternal antibody production) is not recom- contain alloantibodies to antigens on the platelets or platelet
mended because of potential fetal toxicity. In situations in membranes of the transfused blood product, directed against
which the diagnosis of NAIT is known or highly suspected, an antigen the recipient does not have. In more than 90% of
delivery should be by cesarean section to avoid fetal trauma cases, the antibody is directed against the HPA-1a antigen; in
associated with vaginal delivery. After delivery, the affected most of the remaining cases, the antibodies are directed against
infant may be treated with transfusion of the appropriate PlA2 or epitopes on GP IIb/IIIa.37 Involvement of other alloan-
antigen-negative platelets (usually maternal). In addition, tigens, such as HPA-3a (Bak), HPA-4 (Penn), and HPA-5b (Br),
IVIG can be used alone or in combination with platelet transfu- has been reported. The mechanism by which the recipients
sion. IVIG should not be used as the sole treatment in a bleed- own platelets are destroyed is unknown. Most patients with
ing infant, because response to this therapy usually takes 1 to this type of thrombocytopenia are multiparous middle-aged
3 days.63 women. Almost all the other patients have a history of blood
transfusion. PTP seems to be exceedingly rare in men who have
Neonatal Autoimmune Thrombocytopenia. The diag- never been transfused and in women who have never been
nosis of ITP or systemic lupus erythematosus in the mother is pregnant or never been transfused.69 PTP seems to require prior
a prerequisite for the diagnosis of neonatal autoimmune exposure to foreign platelet antigens and behaves in many
thrombocytopenia. Neonatal autoimmune thrombocytopenia respects like an anamnestic immune response.
is due to passive transplacental transfer of antibodies from a No clinical trials have been conducted on the treatment of
mother with ITP or, occasionally, systemic lupus erythemato- PTP, primarily because of the small number of cases. If PTP is
sus. The neonate does not have an ongoing autoimmune untreated or treatment is ineffective, mortality rates may
process per se, but rather is an incidental target of the mothers approach 10%.70 In addition, untreated or unresponsive
autoimmune process. During pregnancy, relapse is relatively patients have a protracted clinical course, with thrombocyto-
common for women with ITP in complete or partial remission; penia typically lasting 3 weeks but in some cases up to 4
this has been attributed to the facilitation of reticuloendothe- months. Plasmapheresis and exchange transfusion have been
lial phagocytosis by the high estrogen levels in pregnant used with some success in the past, but the treatment of choice
women. Women commonly develop chronic ITP during preg- is now IVIG. Many patients with PTP respond to a 2-day course
nancy. ITP in the mother tends to remit after delivery. Cortico- of IVIG, generally within the first 2 to 3 days, although a second
steroids are the primary treatment for pregnant women with course occasionally is necessary.70 IVIG also is much easier to
ITP, and at the dosages used, there is a relatively low incidence administer, and the response rates are higher than for plasma-
of adverse fetal side effects.67 Neonatal autoimmune thrombo- pheresis or exchange transfusion. Corticosteroid therapy is not
cytopenia develops in only about 10% of the infants of particularly efficacious when used alone but may be beneficial
pregnant women with autoimmune thrombocytopenia, and in combination with other, more effective treatments.37
intracranial hemorrhage occurs in 1% or fewer. It is no longer
recommended that high-risk infants be delivered by cesarean Secondary Thrombocytopenia, Presumed to Be
section to avoid the trauma of vaginal delivery and accompany- Immune Mediated. Severe thrombocytopenia has been
ing risk of hemorrhage in the infant, regardless of maternal observed in patients receiving biologic response modifiers such
platelet count.68 as interferons, colony-stimulating factors, and interleukin-2.71-73
Affected newborns may have normal to decreased platelet The thrombocytopenia associated with use of these substances
numbers at birth and have a progressive decrease in the platelet is reversible and, at least for interferon, may be immune medi-
count for about 1 week before the platelet count begins to ated, because increased levels of platelet-associated IgG have
increase. It has been speculated that the falling platelet count been measured. Immune thrombocytopenia develops in about
is associated with maturation of the infants reticuloendothe- 5% to 10% of patients with chronic lymphocytic leukemia and
lial system and accelerated removal of antibody-labeled in a smaller percentage of patients with other lymphoprolifera-
platelets by cells of the reticuloendothelial system. Neonatal tive disorders.74,75 Thrombocytopenia also is noted in 14% to
thrombocytopenia typically persists for about 1 to 2 weeks 26% of patients with systemic lupus erythematosus.76 The clini-
but sometimes lasts for several months. It usually does not cal picture is similar to that of ITP: The bone marrow has a
require treatment. Severely thrombocytopenic infants generally larger than normal number of megakaryocytes, and increased
respond quickly to IVIG treatment. If an infant develops hem- levels of platelet-associated IgG frequently are found.36 Para-
orrhagic symptoms, platelet transfusion, IVIG treatment, or sitic infections also are known to cause thrombocytopenia.
corticosteroid therapy should be started immediately.37 Malaria is the most studied in this group and is regularly
accompanied by thrombocytopenia, the onset of which cor-
Posttransfusion Purpura. Posttransfusion purpura responds to the first appearance of antimalarial antibodies, a
(PTP) is a relatively rare disorder that typically develops about decrease in serum complement, and control of parasitemia.
1 week after transfusion of platelet-containing blood products, There is evidence for the adsorption of microbial antigens to
including fresh or frozen plasma, whole blood, and packed or the platelet surface and subsequent antibody binding via the
706 PART VIII Hemostasis and Thrombosis

Fab terminus.77 Immune destruction of platelets seems to be activation, because low-dose aspirin therapy has been shown
the most likely mechanism for the thrombocytopenia. to prevent preeclampsia in high-risk patients.85,86 When aspirin
is used to prevent preeclampsia, however, reduction in risk is
Nonimmune Mechanisms of Platelet Destruction only 15%.
Nonimmune platelet destruction may result from exposure of The treatment of preeclampsia is delivery of the infant
platelets to nonendothelial surfaces, from activation of the whenever possible. After delivery, the thrombocytopenia
coagulation process, or from platelet consumption by endovas- usually resolves in a few days. In cases in which delivery is not
cular injury without measurable depletion of coagulation possible (e.g., the infant would be too premature), bed rest and
factors.36 aggressive treatment of the hypertension may help to increase
the platelet count in some patients. Such treatments include
Thrombocytopenia in Pregnancy and Preeclampsia magnesium sulfate and other antiepileptic therapies to inhibit
Incidental thrombocytopenia of pregnancy. Incidental eclamptic seizures.
thrombocytopenia of pregnancy also is known as pregnancy-
associated thrombocytopenia and gestational thrombocytopenia. Hemolytic Disease of the Newborn. Thrombocytope-
This disorder is the most common cause of thrombocytopenia nia, usually moderate in degree, occurs frequently in infants
in pregnancy. Random platelet counts in pregnant and post- with hemolytic disease of the newborn. Although the RBC
partum women are slightly higher than normal, but about 5% destruction characteristic of this disorder is antibody induced,
of pregnant women develop a mild thrombocytopenia (100,000 the antigens against which the antibodies are directed are not
to 150,000/mcL), with 98% of such women having platelet expressed on platelets. Platelets may be destroyed as a result of
counts greater than 70,000/mcL. These women are healthy and their interaction with products of RBC breakdown, rather than
have no prior history of thrombocytopenia. They do not seem their direct participation in an immunologic reaction.36
to be at increased risk for bleeding or for delivery of infants Other causes of thrombocytopenia during pregnancy.
with neonatal thrombocytopenia. The cause of this type of As has been discussed previously, ITP is a relatively common
thrombocytopenia is unknown. Maternal platelet counts return disorder in women of childbearing age, and pregnancy does
to normal within several weeks of delivery. These women com- nothing to ameliorate the symptoms of this disorder. ITP
monly experience recurrence in subsequent pregnancies. should be a part of the differential diagnosis of thrombocyto-
Preeclampsia and other hypertensive disorders of penia in a pregnant woman. There is little or no correlation
pregnancy. Approximately 20% of cases of thrombocytope- between the level of maternal autoantibodies and the fetal
nia of pregnancy are associated with hypertensive disorders. platelet count. Other causes of thrombocytopenia during preg-
These disorders include the classifications preeclampsia, nancy include HIV infection, systemic lupus erythematosus,
preeclampsia-eclampsia, preeclampsia with chronic hyperten- antiphospholipid syndromes, TTP, and HUS. Of all women
sion, chronic hypertension, and gestational hypertension. who develop TTP, 10% to 25% manifest the disease during
Preeclampsia complicates about 5% of all pregnancies and pregnancy or in the postpartum period, and TTP tends to recur
typically occurs at about 20 weeks of gestation. The disorder is in subsequent pregnancies.87,88 Plasmapheresis is the treatment
characterized by the onset of hypertension and proteinuria and of choice, and the maternal mortality is 90% or greater without
may include abdominal pain, headache, blurred vision, or such treatment.
mental function disturbances.78 Thrombocytopenia occurs in
15% to 20% of patients with preeclampsia, and about 40% to Thrombotic Thrombocytopenic Purpura. TTP, some-
50% of these patients progress to eclampsia (hypertension, times referred to as Moschcowitz syndrome, is characterized by
proteinuria, and seizures).79,80 the triad of microangiopathic hemolytic anemia, thrombocy-
Some patients with preeclampsia have microangiopathic topenia, and neurologic abnormalities.89 In addition, fever and
hemolysis, elevated liver enzymes, and a low platelet count, renal dysfunction (forming a pentad) are often present. Addi-
termed HELLP syndrome. HELLP syndrome affects an estimated tional symptoms are present in most patients at the time of
4% to 12% of women with severe preeclampsia45,81,82 and diagnosis and include diarrhea, anorexia, nausea, weakness,
seems to be associated with higher rates of maternal and fetal and fatigue. TTP is uncommon but not rare, and its incidence
complications. This disorder may be difficult to differentiate may be increasing. About twice as many women as men are
from thrombotic thrombocytopenic purpura (TTP), hemolytic affected, and it is most common in women 30 to 40 years of
uremic syndrome (HUS), and disseminated intravascular age.34,90 About half of the patients who develop TTP have a
coagulation (DIC). history of a viral-like illness several days before the onset
The development of thrombocytopenia in these patients of TTP.
is thought to be due to increased platelet destruction. The There seem to be at least four types of TTP. In most patients,
mechanism of platelet destruction is unclear, however. Some TTP occurs as a single acute episode, although a small fraction
evidence (elevated D-dimer) suggests that these patients have of these patients may have recurrence at seemingly random
an underlying low-grade DIC.83 Elevated platelet-associated intervals. Recurrent TTP occurs in 11% to 28% of TTP
immunoglobulin is commonly found in these patients, patients.91,92 A third type of TTP is drug induced. The primary
however, which indicates immune involvement.84 Early reports agents involved are the purinoreceptor (adenosine diphos-
suggested that there may be a component of in vivo platelet phate) blocking agents ticlopidine (Ticlid) and clopidogrel
CHAPTER 43 Thrombocytopenia and Thrombocytosis 707

(Plavix) used for inhibition of platelet function. Ticlopidine


seems to cause TTP in about 0.025% of patients, whereas the
incidence of clopidogrel-induced TTP is approximately four
times less frequent.90 Another type is chronic relapsing TTP, a
rare form of TTP in which episodes occur at intervals of approx-
imately 3 months starting in infancy.93,94
Although it is unclear what triggers their deposition, hyaline
thrombi are found in the end arterioles and capillaries. These
hyaline thrombi are composed of platelets and von Willebrand
factor (VWF) but contain very little fibrin or fibrinogen. As
these platelet-VWF thrombi are deposited, thrombocytopenia
develops. The degree of thrombocytopenia is directly related
to the extent of microvascular platelet aggregation. RBCs
flowing under arterial pressure are prone to fragmentation and
hemolysis when they encounter the strands of these thrombi. A
Hemolysis is usually quite severe, and most patients have
less than 10g/dL hemoglobin at the time of diagnosis. Exami-
nation of the peripheral blood film reveals a marked decrease
in platelets, RBC polychromasia, and RBC fragmentation
(microspherocytes, schistocytes, keratocytes), a triad of features
characteristic of microangiopathic hemolytic anemias (Figure
43-8). Nucleated RBC precursors also may be present depend-
ing on the degree of hemolysis. Other laboratory evidence of
intravascular hemolysis includes reduction of haptoglobin,
hemoglobinuria, hemosiderinuria, increased serum unconju-
gated bilirubin, and increased lactate dehydrogenase activity.
Bone marrow examination reveals erythroid hyperplasia and a
normal to increased number of megakaryocytes. The partial
thromboplastin time, prothrombin time, fibrinogen, fibrin
degradation products, and D-dimer test results are usually B
normal and may be useful in differentiating this disorder
from DIC. Figure 43-8 Microangiopathic hemolytic anemia in thrombotic thrombo-
The thrombotic lesions also give rise to the other character- cytopenia purpura. Abundant schistocytes reflect platelet clots in the micro-
vasculature. (From Carr JH, Rodak BF: Clinical hematology atlas, ed 3,
istic manifestations of TTP, because they are deposited in the
Philadelphia, 2009, Saunders.)
vasculature of all organs. The thrombi occlude blood flow and
lead to organ ischemia. Symptoms depend on the severity of
ischemia in each organ. Neurologic manifestations range from VWF multimers (ULVWF multimers) than those found in
headache to paresthesia and coma. Visual disturbances may be plasma, and these are even more effective than the normal
of neurologic origin or may be due to thrombi in the choroid plasma VWF multimers at binding platelet GP Ib/IX or GP
capillaries of the retina or hemorrhage into the vitreous. Renal IIb/IIIa complexes under fluid shear stresses (Figure 43-9).93
dysfunction is common and present in more than half of In the plasma, the ULVWF multimers are rapidly cleaved into
patients.91,92 Symptoms of renal dysfunction include protein- the smaller VWF multimers normally found in the plasma
uria and hematuria. Overwhelming renal damage with anuria by a VWF-cleaving metalloprotease, called a disintegrin-like
and fulminant uremia usually does not occur, however, which and metalloprotease domain with thrombospondin type 1 motifs
helps to distinguish TTP from HUS.10 Gastrointestinal bleeding (ADAMTS 13). The metalloprotease seems to be more effective
occurs frequently in severely thrombocytopenic patients, and when VWF multimers are partially unfolded by high shear
abdominal pain is occasionally present due to occlusion of the stress.95,96
mesenteric microcirculation. Familial chronic relapsing TTP is a form characterized by
ADAMTS 13 and thrombotic thrombocytopenic pur recurrent episodes of thrombocytopenia with or without isch-
pura. The development of TTP seems to be directly related emic organ damage. In this type of TTP, the VWF-cleaving
to the accumulation of unusually large von Willebrand factor metalloprotease is completely deficient. The more common
(ULVWF) multimers in the plasma of patients with TTP. VWF form of TTP (usually not familial) does not tend to recur, but
multimers are made by megakaryocytes and endothelial cells. patients also are deficient in the metalloprotease. In this more
The primary source of plasma VWF seems to be endothelial common form of TTP, the metalloprotease deficiency occurs
cells. Endothelial cells secrete VWF into the subendothelium through removal of the enzyme (or blockade of its function)
and plasma and store it in Weibel-Palade bodies (storage gran- by a specific autoantibody that is present during TTP but
ules). Endothelial cells and megakaryocytes make even larger disappears during remission.97,98 An assay to measure the
708 PART VIII Hemostasis and Thrombosis

+ADAMTS 13 Normal VWF


Normal VWF multimers
Spontaneous Unusually large VWF
multimer
synthesis multimers

ADAMTS 13
Platelet agglutination
and aggregation

Figure 43-9 Mechanism for thrombotic thrombocytopenia purpura (TTP). Unusually large von Willebrand factor (ULVWF) multimers are normally digested
by the VWFcleaving protease ADAMTS 13 (see text for full name). In TTP, the absence of ADAMTS 13 allows the release of ULVWF, triggering platelet
activation.

VWF-cleaving enzyme has been introduced.99 However, the test prostacyclin, heparin, or fibrinolytic agents is controversial and
may take several days and treatment decisions must usually be has not clearly been shown to be helpful. Platelet transfusions
made before test results are available. In addition, the assay should be avoided unless intracranial bleeding or other serious
lacks sensitivity and specificity for TTP. Additional tests for hemorrhagic problems arise.45
ADAMTS 13 (and TTP) are in development and hold the Before 1990, TTP was fatal in more than 80% of patients.
promise for rapid diagnosis of TTP. With the means for rapid diagnosis and the advent of exchange
In patients with TTP, ULVWF multimers tend to be present plasmapheresis, now 80% of patients who are treated early can
in the plasma at the beginning of the episode. These ULVWF be expected to survive. Because patients are known to experi-
multimers, and usually the normal-sized plasma multimers, ence relapse, however, platelet counts should be monitored on
disappear as the TTP episode progresses and the thrombocyto- a regular basis until patients are in remission. The detection of
penia worsens. Platelets and VWF are consumed during the ULVWF multimers in patient samples after complete remission
deposition of the microvascular hyaline thrombi characteristic has predicted relapse accurately in 90% of the patients tested102;
of this disorder.100 If the patient survives an episode of TTP and this may prove to be useful in the long-term management
does not experience relapse, the plasma VWF multimers are of TTP.
usually normal after recovery. If ULVWF multimers are found
in the plasma after recovery, however, it is likely that the Hemolytic Uremic Syndrome. Clinically, HUS resem-
patient will have recurrent episodes of TTP. The episodes may bles TTP except that it is found predominantly in children 6
be infrequent and at irregular intervals (intermittent TTP) or months to 4 years of age and is self-limiting. Approximately
frequent and at regular intervals, as is often the case when TTP 90% of cases are caused by Shigella dysenteriae serotypes or
episodes occur in early childhood or infancy. enterohemorrhagic Escherichia coli OH serotypes, particularly
The most effective treatment for TTP is plasma exchange O157:H7.103 These organisms sometimes can be cultured from
using fresh frozen plasma or cryoprecipitate-poor plasma stool samples. The bloody diarrhea typical of childhood HUS
(plasma lacking most of the fibrinogen, fibronectin, and is caused by colonization of the large intestine with the offend-
VWF).101 Either of these approaches may produce dramatic ing organism, which causes erosive damage to the colon. S.
effects within a few hours. Because plasmapheresis is not avail- dysenteriae produces Shiga toxin, and enterohemorrhagic E. coli
able in all centers, the patient should be given corticosteroids produce either Shiga-like toxin 1 (SLT-1) or SLT-2, which can
and infusions of fresh frozen plasma immediately. Plasma be detected in fecal samples from patients with HUS. The
exchange should be arranged as quickly as possible. Plasma- toxins enter the bloodstream and attach to renal glomerular
pheresis and replacement/infusion of plasma is effective on capillary endothelial cells, which become damaged and swollen
two fronts. First, some of the ULVWF multimers will be and release ULVWF multimers.103,104 This process leads to for-
removed by apheresis, and plasma (fresh frozen plasma or mation of hyaline thrombi in the renal vasculature and the
cryoprecipitate-poor plasma) supplies the deficient protease, development of renal failure, thrombocytopenia, and micro
which is able to degrade the ULVWF multimers in the blood angiopathic hemolytic anemia, although the RBC fragmenta-
of the patient. Because some patients with TTP have recovered tion is usually not as severe as that seen in TTP (this is, however,
while receiving immunosuppressive treatment (corticoste- not a differentiating feature). The extent of renal involvement
roids) alone, it is recommended that all patients with TTP be correlates with the rate of recovery. In more severely affected
given high-dose corticosteroids in addition to undergoing children, renal dialysis may be needed. The mortality rate asso-
plasma exchange. Plasma exchange typically is continued over ciated with HUS in children is much lower than that for TTP,
a 5-day period. If a response is not seen within 5 days, or if the but there is often residual renal dysfunction that may lead to
condition of the patient worsens during the first few days of renal hypertension and severe renal failure. Because HUS in
plasma exchange, additional treatment is instituted. Such ther- children is essentially an infectious disorder, it affects boys and
apies include administering vincristine or azathioprine (or girls equally and is often found in geographic clusters of cases
other immunosuppressive agents), performing splenectomy, rather than in random distribution.
or passing the patients plasma over a staphylococcal A column The adult form of HUS is associated most often with expo-
to remove immune complexes. The use of antiplatelet agents, sure to immunosuppressive agents or chemotherapeutic agents
CHAPTER 43 Thrombocytopenia and Thrombocytosis 709
or both, but it also may occur during the postpartum period. In chronic DIC, there is an ongoing, low-grade consumptive
Usually the symptoms of HUS do not appear until weeks or coagulopathy. Clotting factors may be slightly reduced or
months after exposure to the offending agent.105 This disorder normal, and compensatory thrombocytopoiesis results in a
most likely results from direct renal arterial endothelial damage moderately low to normal platelet count.34 D-dimer may not
caused by the drug or one of its metabolic products. The be detectable or may be slightly to moderately increased.
damage to endothelial cells results in release of VWF (including Chronic DIC is not generally life-threatening, and treatment
ULVWF multimers), turbulent flow in the arterial system with usually is not urgent. Chronic DIC is almost always due to
increased shear stresses on platelets, and VWF-mediated plate- some underlying condition. If that condition can be corrected,
let aggregation in the renal arterial system. The renal impair- the DIC usually resolves without further treatment. Chronic
ment in adults seems to be more severe than that in childhood DIC should be followed closely, however, because it can
HUS, and dialysis is usually required. The cause of HUS associ- convert into the life-threatening acute form.
ated with pregnancy or oral contraceptive use is unclear, but it
may be related to development of an autoantibody to endo- Drug-Induced Thrombocytopenia. A few drugs
thelial cells. In outbreaks of HUS associated with consumption directly interact with platelets to cause thrombocytopenia. Ris-
of E. colicontaminated water, both children and adults have tocetin, an antibiotic no longer in clinical use, facilitates the
developed HUS. interaction of VWF with platelet membrane GP Ib and leads to
The cardinal signs of HUS are hemolytic anemia, renal in vivo platelet agglutination and thrombocytopenia. Hematin,
failure, and thrombocytopenia. The thrombocytopenia is used for the treatment of acute intermittent porphyria, may
usually mild to moderate in severity. Renal failure is reflected give rise to a transient thrombocytopenia that seems to be
in elevated blood urea nitrogen and creatinine levels. The urine caused by stimulation of platelet secretion and aggregation.
nearly always contains RBCs, protein, and casts. The hemolytic Protamine sulfate and bleomycin may induce thrombocytope-
process is shown by a hemoglobin level of less than 10g/dL, nia by a similar mechanism.45
elevated reticulocyte count, and presence of schistocytes in the
peripheral blood. Abnormalities in Distribution or Dilution
Differentiating the adult form of HUS from TTP may be An abnormal distribution of platelets also may cause throm-
difficult. The lack of neurologic symptoms, the presence bocytopenia. The normal spleen sequesters approximately one
of renal dysfunction, and the absence of other organ involve- third of the total platelet mass. Mild thrombocytopenia may
ment suggest HUS. Also, in HUS the thrombocytopenia tends be present in any of the big spleen syndromes. The total body
to be mild to moderate (platelet consumption occurs primarily platelet mass is often normal in these disorders, but numerous
in the kidneys), whereas in TTP the thrombocytopenia is platelets are sequestered in the enlarged spleen, and conse-
usually severe. Similarly, fragmentation of RBCs and the resul- quently the venous blood platelet count is low. Disorders such
tant anemia tend to be milder than that observed in TTP, as Gaucher disease, Hodgkin disease, sarcoidosis, lymphoma,
because RBCs are being fragmented primarily in the kidneys. cirrhosis of the liver, and portal hypertension may result in
In some cases of HUS, other organs become involved, and splenomegaly and lead to thrombocytopenia.
the differentiation between HUS and TTP becomes less clear. Lowering the body temperature to less than 25 C, as is
In such cases, it is prudent to treat as though the patient routinely done in cardiovascular surgery, results in a transient
has TTP. but mild thrombocytopenia secondary to platelet sequestra-
tion in the spleen and liver. An associated transient defect in
Disseminated Intravascular Coagulation. A common function also occurs with hypothermia. Platelet count and
cause of destructive thrombocytopenia is activation of the function return to baseline values on return to normal body
coagulation cascade (by a variety of agents or conditions), temperature.32
resulting in a consumptive coagulopathy that entraps platelets Thrombocytopenia often follows surgery involving extra-
in intravascular fibrin clots. This disorder is described in more corporeal circulatory devices, as a consequence of damage and
detail in Chapter 42 but is discussed here briefly for the sake partial activation of platelets in the pump. In a few cases, severe
of completeness. DIC has many similarities to TTP, including thrombocytopenia, marked impairment of platelet function,
microangiopathic hemolytic anemia and deposition of thrombi and activation of fibrinolysis and intravascular coagulation
in the arterial circulation of most organs. In DIC, however, the may develop.45
thrombi are composed primarily of platelets and fibrinogen, The administration of massive amounts of stored whole
whereas in TTP the thrombi are composed primarily of plate- blood may produce a temporary thrombocytopenia. This phe-
lets and VWF. nomenon is explained by the fact that stored blood contains
One form of DIC is acute with rapid platelet consumption platelets whose viability is severely impaired by the effects of
and results in severe thrombocytopenia. In addition, levels of storage and temperature. Under these conditions, the dead or
factor V, factor VIII, and fibrinogen are decreased as a result of damaged platelets are rapidly sequestered by the reticuloendo-
in vivo thrombin generation. The test for D-dimer (a break- thelial system of the patient. If this problem is encountered, it
down product of stabilized fibrin) almost always yields posi- may be minimized by administering platelet concentrates or
tive results. This form of DIC is life-threatening and must be units of fresh whole blood along with the stored blood. This
treated immediately. situation is only rarely encountered, however, because the
710 PART VIII Hemostasis and Thrombosis

practice of transfusing whole blood has been replaced almost malignancy. Occasionally, patients manifest a platelet count of
completely by the use of specific components. Finally, mild 1 to 2 million/mcL (see Figure 43-10). In reactive thrombocy-
thrombocytopenia may be encountered in patients with tosis, platelet production remains responsive to normal regula-
chronic renal failure, severe iron deficiency, megaloblastic tory stimuli (e.g., thrombopoietin, a humoral factor that is
anemia, postcompression sickness, and chronic hypoxia. produced in the kidney parenchyma), and morphologically
normal platelets are produced at a moderately increased rate.
This is in contrast to essential thrombocythemia, which is char-
THROMBOCYTOSIS: INCREASE
acterized by unregulated or autonomous platelet production
IN CIRCULATING PLATELETS
and platelets of variable size.106,107
Thrombocytosis is defined as an abnormally high platelet Examination of the bone marrow from patients with reac-
count, typically more than 450,000/mcL. The term reactive tive thrombocytosis reveals a normal to increased number of
thrombocytosis is used to describe an elevation in the platelet megakaryocytes that are normal in morphology. Results of
count secondary to inflammation, trauma, or other underlying platelet function tests, including aggregation induced by
and seemingly unrelated conditions. In reactive thrombocyto- various agents, and bleeding time are usually normal in
sis, the platelet count is elevated for a limited period and
usually does not exceed 800,000/mcL, although platelet counts
greater than 1 million/mcL are occasionally seen (Figure
43-10). A marked and persistent elevation in the platelet count BOX 43-3 Processes Resulting in Thrombocytosis
is a hallmark of myeloproliferative disorders such as polycy-
themia vera, chronic myelogenous leukemia, and myelofibro- Conditions Associated with Reactive
sis with myeloid metaplasia. In these conditions, the platelet Thrombocytosis
count often exceeds 1 million/mcL. Although the terms throm- Blood loss and surgery
bocythemia and thrombocytosis are often used interchangeably, Splenectomy
in this text the term thrombocythemia is used only as part of the Iron deficiency anemia
description of the myeloproliferative disorder known as essen- Inflammation and disease
tial thrombocythemia (Figure 43-11). In essential thrombocythe- Stress or exercise
mia, platelet counts typically exceed 1 million/mcL and may
reach levels of several million.34,106,107 Processes resulting in Myeloproliferative Disorders Associated with
thrombocytosis are summarized in Box 43-3. Thrombocytosis
Polycythemia vera
Reactive (Secondary) Thrombocytosis Chronic myelogenous leukemia
Platelet counts between 450,000/mcL and 800,000/mcL with Myelofibrosis with myeloid metaplasia
no change in platelet function can result from acute blood loss, Thrombocythemia: essential or primary
splenectomy, childbirth, and tissue necrosis secondary to Data from Colvin BT: Thrombocytopenia, Clin Haematol 14:661-681, 1985;
surgery, chronic inflammatory disease, infection, exercise, iron and Thompson AR, Harker LA: Manual of hemostasis and thrombosis, ed 3,
deficiency anemia, hemolytic anemia, renal disorders, and Philadelphia, 1983, FA Davis.

Figure 43-11 Peripheral blood film showing cell morphology in essential


thrombocythemia. Note the increased number of platelets and wide variation
Figure 43-10 Peripheral blood film showing cell morphology in reactive in platelet size characteristic of essential thrombocythemia. Red and white
thrombocytosis. Note the increased number of platelets but reasonably blood cell morphology is characteristically normal. (From Carr JH, Rodak BF:
normal platelet morphology, characteristic of reactive thrombocytosis. Clinical hematology atlas, ed 3, Philadelphia, 2009, Saunders.)
CHAPTER 43 Thrombocytopenia and Thrombocytosis 711
reactive thrombocytosis but also may be normal in patients association with rheumatoid arthritis, rheumatic fever, osteo-
with elevated platelet counts accompanying myeloproliferative myelitis, ulcerative colitis, acute infections, and malignancy. In
disorders. rheumatoid arthritis, the presence of thrombocytosis can be
Reactive thrombocytosis is not associated with thrombosis, correlated with activation of the inflammatory process.
hemorrhage, or abnormal thrombopoietin levels. It seldom Kawasaki disease (Kawasaki syndrome) is an acute febrile
produces symptoms per se and disappears when the underly- illness of infants and young children. It is a self-limited acute
ing disorder is brought under control.106,107 vasculitic syndrome of unknown origin, although an infectious
etiology has been suspected. Although the disease is self-
Reactive Thrombocytosis Associated with limiting, there can be lifelong sequelae, including coronary
Hemorrhage or Surgery artery thrombosis and aneurysms. The acute febrile stage of the
After acute hemorrhage, the platelet count may be low for 2 to disease lasts 2 weeks or longer with fever of 40 C or higher
6 days (unless platelets have been transfused) but typically and is unresponsive to antibiotic therapy. The longer the fever
rebounds to elevated levels for several days before returning to continues, the higher the risk of cardiovascular complications.
the prehemorrhage level. A similar pattern of thrombocytope- The subacute phase lasts an additional week to 10 days. During
nia and thrombocytosis is seen after major surgical procedures this phase, the platelet count usually is elevated, and counts of
in which there is significant blood loss. In both cases, the 2 million/mcL have been reported. In addition, acute phase
platelet count typically returns to normal 10 to 16 days after reactants such as C-reactive protein and sedimentation rate are
the episode of blood loss. elevated. The WBC count can be moderately to markedly
elevated with a left shift, and many patients develop a mild
Postsplenectomy Thrombocytosis normochromic, normocytic anemia. During this phase, cardio-
Removal of the spleen typically results in platelet counts that vascular complications and aneurysms develop. The higher the
can reach or exceed 1 million/mcL regardless of the reason for platelet count, the higher the risk of cardiovascular complica-
splenectomy. The spleen normally sequesters about one third tion. After the subacute phase comes the convalescent phase,
of the circulating platelet mass. After splenectomy, one would during which all signs of illness disappear and the acute phase
expect an initial increase in the platelet count of approximately reactants subside to normal. The highest incidence of Kawasaki
30% to 50%. The platelet count, however, far exceeds levels disease is found in Japan and in individuals of Japanese
that could result from rebalancing of the circulating platelet descent, although the disease seems to occur in most, if not all,
pool to incorporate the splenic platelet pool. The cause of the ethnic groups. The treatment for Kawasaki disease is admini
accelerated platelet production is unknown. Unlike after blood stration of antiplatelet agents and immunoglobulin.
loss from hemorrhage or other types of surgery, the platelet An elevated platelet count also may be early evidence of a
count reaches a maximum 1 to 3 weeks after splenectomy and tumor (e.g., Hodgkin disease) and various carcinomas. Finally,
remains elevated for 1 to 3 months. In some patients who hemophilic patients often have platelet counts above normal
undergo splenectomy for treatment of chronic anemia, the limits, even in the absence of active bleeding.
count can remain elevated for several years.
Exercise-Induced Thrombocytosis
Thrombocytosis Associated with Strenuous exercise is a well-known cause of relative thrombo-
Iron Deficiency Anemia cytosis and is likely due to the release of platelets from the
Mild iron deficiency anemia secondary to chronic blood loss splenic pool or hemoconcentration by transfer of plasma water
is associated with thrombocytosis in about 50% of cases. to the extravascular compartment or both. Normally, the plate-
Thrombocytosis can be seen in severe iron deficiency anemia, let count returns to its preexercise baseline level 30 minutes
but thrombocytopenia also has been reported. In some cases after completion of exercise.
of iron deficiency, the platelet count may be 2 million/mcL.
After iron therapy is started, the platelet count usually returns Rebound Thrombocytosis
to normal within 7 to 10 days. It is believed that iron plays Thrombocytosis often follows the thrombocytopenia caused
some role in regulating thrombopoiesis, because treatment of by marrow-suppressive therapy or other conditions. Rebound
the iron deficiency with iron replacement has resulted in a thrombocytosis usually reaches a peak 10 to 17 days after
normalization of the platelet count in thrombocytopenic withdrawal of the offending drug (e.g., alcohol or methotrex-
patients and has been reported to induce thrombocytopenia ate) or after institution of therapy for the underlying condition
in patients with normal platelet counts. Not enough research with which thrombocytopenia is associated (e.g., vitamin B12
has been done, however, to elucidate the role of iron in deficiency).45
thrombopoiesis.
Thrombocytosis Associated with
Thrombocytosis Associated with Inflammation Myeloproliferative Disorders
and Disease Primary or autonomous thrombocytosis is a typical finding in
Similar to elevations in C-reactive protein, fibrinogen, VWF, four chronic myeloproliferative disorders: polycythemia vera,
and other acute phase reactants, thrombocytosis may be an chronic myelogenous leukemia, myelofibrosis with myeloid
indication of inflammation. Thrombocytosis may be found in metaplasia, and essential thrombocythemia. Depending on the
712 PART VIII Hemostasis and Thrombosis

duration and stage of the myeloproliferative disorder at the and -thromboglobulin in the blood. These findings suggest
time of diagnosis, it may be difficult to differentiate among enhanced in vivo platelet activation and perhaps an explana-
these diseases. Chapter 34 provides a more complete descrip- tion for the thrombotic tendencies of patients with essential
tion of these disorders. In the other types of myeloproliferative thrombocythemia. The primary cause of death of patients with
disorders, the platelet count seldom reaches the extreme essential thrombocythemia seems to be thrombosis. Hemor-
values characteristic of essential thrombocythemia. Diagnosis rhagic episodes occur less frequently than thrombotic episodes
of essential thrombocythemia should not be based on in patients with essential thrombocythemia.
the platelet count alone but should also take into account As with bleeding secondary to platelet function disorders,
the physical examination findings, history, and other labora- the hemorrhagic manifestations of essential thrombocythemia
tory data.34,107 are mucocutaneous in nature, with gastrointestinal tract bleed-
ing occurring most frequently. Other sites of bleeding include
Essential (Primary) Thrombocythemia the mucous membranes of the nose and mouth, the urinary
Essential thrombocythemia is a clonal disorder related to other tract, and the skin. Symptoms may be aggravated by aspirin
chronic myeloproliferative diseases and is the most common use. In most cases of thrombocythemia, patients experience no
cause of primary thrombocytosis. It is characterized by periph- clotting disorders. In an occasional patient with essential
eral blood platelet counts exceeding 1 million/mcL and uncon- thrombocythemia, there is a paradoxical combination of
trolled proliferation of marrow megakaryocytes. Although the thromboembolic (clotting) and hemorrhagic episodes in
platelet count may (or may not) be markedly elevated in other association with this condition. A patient with essential throm-
myeloproliferative disorders, persistent marked elevation of bocythemia who has had a thrombotic event may have a
the platelet count is an absolute requirement for the diagnosis hemorrhagic event later.110
of essential thrombocythemia (see Figure 43-11). There is evi- The bleeding manifestations may be related to a variety of
dence that essential thrombocythemia is caused by a clonal qualitative abnormalities in the platelets, including deficien-
proliferation of a single abnormal pluripotential stem cell that cies in epinephrine receptors and ultrastructural defects in
eventually crowds out normal stem cells. As with most myelo- granules, mitochondria, and microfilaments. Platelets may be
proliferative disorders, essential thrombocythemia is neither agranular or hypogranular and have a clear, light blue appear-
congenital nor hereditary, is prevalent in middle-aged and ance on a routine Wright-stained film of the peripheral blood.
older patients, and affects equal numbers of men and women. Although platelet size is heterogeneous, giant and bizarrely
In contrast to other myeloproliferative disorders, however, shaped platelets are characteristic of myeloproliferative dis-
the other marrow cell lines are not involved at the time of eases, and megakaryocyte fragments or nuclei are commonly
diagnosis. encountered in the peripheral blood. Platelets may be notably
The clinical manifestations of essential thrombocythemia clumped on blood films, exhibiting marked variation in size
are hemorrhage, platelet dysfunction, and thrombosis. Bleed- and shape. The number and volume of megakaryocytes are
ing times are usually normal. There is no specific clinical sign, increased in the bone marrow, and they are predominantly
symptom, or laboratory test that establishes the diagnosis of large, show some cellular atypia, and tend to form clusters.
essential thrombocythemia. The diagnosis must be made by Often the platelets are functionally defective when tested in
ruling out the other myeloproliferative disorders and systemic vitro. Aggregation is usually absent in response to epinephrine
illnesses that produce reactive thrombocytosis. and may be decreased with adenosine diphosphate but is
Thrombosis in the microvasculature is relatively common usually normal with collagen. Lack of an epinephrine response
in essential thrombocythemia, and the incidence at the time may help to differentiate essential thrombocythemia from
of diagnosis is 10% to 20%. This thrombosis can lead to digital reactive thrombocytosis, because this response is usually
pain, digital gangrene, or erythromelalgia (throbbing, aching, normal in reactive thrombocytosis but absent in most cases of
and burning sensation in the extremities, particularly in the essential thrombocythemia. Platelet adhesion also may be
palms and soles).107 The symptoms of erythromelalgia can be decreased.107
explained by arteriolar inflammation and occlusive thrombosis The degree of thrombocytosis has not been found to predict
mediated by platelets and can be relieved for several days by a hemorrhagic or thrombotic events reliably. The role of lower-
single dose of aspirin.108 Thrombosis of large veins and arteries ing platelet counts as a prophylactic treatment in this disease
also may occur in essential thrombocythemia. The arteries is not established. The risks from exposure to mutagenic alkyl-
most commonly involved are those in the legs, the coronary ating agents used to decrease the platelet count may be greater
arteries, and the renal arteries, but involvement of the mesen- than the risk of thrombosis or hemorrhage. When treatment is
teric, subclavian, and carotid arteries is not infrequent (in fact, deemed necessary because of thrombotic tendencies or spleno-
neurologic complications are relatively common). Venous megaly, a variety of myelosuppressive agents (e.g., melphalan,
thrombosis may involve the large veins of the legs and busulfan, hydroxyurea, or even radioactive phosphorus) can be
pelvis, hepatic veins, or splenic veins.109 The platelets of some used.107 In patients with life-threatening hemorrhage or throm-
patients who have experienced thrombotic episodes have been bosis and an extremely high platelet count, platelet apheresis
shown to have increased binding affinity for fibrinogen and to may be used to reduce the platelet count rapidly. In these situ-
generate more than the usual quantities of thromboxane B2, ations, a myelosuppressive agent is added for longer-term
and these patients have elevated levels of thromboxane B2 control of the platelet count.111 Interferon- has been used to
CHAPTER 43 Thrombocytopenia and Thrombocytosis 713
treat essential thrombocythemia and is associated with an availability of newer treatments for essential thrombocythe-
approximately 60% rate of complete remission, but 28% of mia, whether a patient with essential thrombocythemia and an
patients given the drug cannot tolerate the dosages required.112-114 elevated platelet count who is asymptomatic should or should
A newer agent that has shown great promise in the treatment not be treated remains controversial.
of essential thrombocythemia is anagrelide. The drug acts by In patients with essential thrombocythemia there is a low
inhibiting megakaryocyte maturation and platelet release.115 In incidence of transformation to acute leukemia or fatal throm-
one large study, anagrelide decreased the platelet count in 93% botic or hemorrhagic complications. Therapy to prevent
of patients.116 Many patients cannot tolerate anagrelide, thrombotic complications seems to be effective in preventing
however, and in these patients other, more traditional chemo- morbidity but does not seem to improve survival, at least in
therapeutic agents seem to be more effective. Despite the high-risk patients.

SUMMARY
Thrombocytopenia is the most common cause of clinically signifi- Treatment of drug-induced thrombocytopenia must begin with
cant bleeding. identification of the causative drug and discontinuation of its use.
Thrombocytopenia may be a result of decreased platelet produc- TTP presents with a triad of symptoms that includes microangio-
tion, increased destruction, or abnormal distribution of platelets pathic hemolytic anemia, thrombocytopenia, and neurologic
and manifests with small-vessel bleeding in the skin. abnormalities; these may be accompanied by fever and renal
Decreased production of platelets can be attributed to megakaryo- dysfunction.
cyte hypoplasia, ineffective thrombopoiesis, or replacement of Abnormal distribution of platelets can be caused by splenic
marrow by abnormal cells. sequestration.
Patients experiencing increased platelet destruction become Reactive thrombocytosis is secondary to inflammation, trauma, or
thrombocytopenic only when the rate of platelet production can a variety of underlying conditions. Platelet counts are increased
no longer increase enough to compensate. for a limited time. The thrombocytosis seen in myeloproliferative
Pathologic destruction of platelets can be caused by both immu- disorders is marked and persistent.
nologic and nonimmunologic mechanisms.
Acute ITP commonly occurs in children after a viral illness, and Now that you have completed this chapter, go back and
there is usually spontaneous remission. Chronic ITP is more com- read again the case study at the beginning and respond
monly seen in women and requires treatment if the platelet count to the questions presented.
decreases to fewer than 30,000/mcL.

R E V I E W Q UESTIONS
1. The autosomal dominant disorder associated with 4. A 2-year-old child with an unexpected platelet count of
decreased platelet production is: 15,000/mcL and a recent history of a viral infection most
a. Fanconi anemia likely has:
b. TAR syndrome a. HIT
c. May-Hegglin anomaly b. NAIT
d. Wiskott-Aldrich anomaly c. Acute ITP
d. Chronic ITP
2. Which of the following is not a hallmark of ITP?
a. Petechiae 5. What is the first step in the treatment of HIT?
b. Thrombocytopenia a. Start low-molecular-weight heparin therapy.
c. Large overactive platelets b. Stop heparin infusion immediately.
d. Megakaryocyte hypoplasia c. Switch to warfarin (Coumadin) immediately.
d. Initiate a platelet transfusion.
3. The specific antigen most commonly responsible for the
development of NAIT is: 6. A defect in primary hemostasis (platelet response to an
a. Bak injury) often results in:
b. HPA-1a a. Musculoskeletal bleeding
c. GP Ib b. Mucosal bleeding
d. Lewis antigen a c. Hemarthroses
d. None of the above
714 PART VIII Hemostasis and Thrombosis

7. When a drug acts as a hapten to induce thrombocytopenia, 9. Neonatal autoimmune thrombocytopenia occurs when:
an antibody forms against which of the following? a. The mother lacks a platelet antigen that the infant
a. Typically unexposed, new platelet antigens possesses, and she builds antibodies to that antigen,
b. The combination of the drug and the platelet which cross the placenta.
membrane protein to which it is bound b. The infant develops an autoimmune process such as
c. The drug alone in the plasma, but the immune ITP secondary to in utero infection.
complex then binds to the platelet membrane c. The infant develops an autoimmune disease such as
d. The drug alone, but only when it is bound to the lupus erythematosus before birth.
platelet membrane d. The mother has an autoimmune antibody to her own
platelets, which crosses the placenta and reacts with
8. TAR refers to: the infants platelets.
a. Abnormal platelet morphology in which the radial
striations of the platelets are missing 10. HUS in children is associated with:
b. Abnormal appearance of the iris of the eye in which a. Diarrhea caused by Shigella species
radial striations are absent b. Meningitis caused by Haemophilus species
c. Abnormal bone formation, including hypoplasia of c. Pneumonia caused by Mycoplasma species
the forearms d. Pneumonia caused by respiratory viruses
d. Neurologic defects affecting the root (radix) of the
spinal nerves

REFERENCES
1. Kelley MJ, Jawien W, Ortel TL, et al: Mutations of MYH9, 13. Freson KK, Devriendt G, Matthijs G, et al: Platelet character-
encoding for non-muscle myosin heavy chain A, in May istics in patients with X-linked macrothrombocytopenia
Hegglin anomaly. Nat Genet 26:106-108, 2000. because of a novel GATA-1 mutation. Blood 98:85-92, 2001.
2. The May Hegglin/Fechtner Syndrome Consortium: Muta- 14. Casteels AA, Naessens F, Cordts L, et al: Neonatal screening
tions in MHY9 result in May-Hegglin anomaly, and for congenital cytomegalovirus infections. J Perinat Med
Fechtner and Sebastian syndromes. Nat Genet 26:103-105, 27:116-121, 1999.
2000. 15. Brown HL, Abernathy MP: Cytomegalovirus infection.
3. Mhawech P, Saleem A: Inherited giant platelet disorders: Semin Perinatol 22:260-266, 1998.
classification and literature review. Am J Clin Pathol 113:176- 16. Crapnell KED, Zanfani A, Chaudhuri JL, et al: In vitro infec-
190, 2000. tion of megakaryocytes and their precursors by human cyto-
4. Symonds RP, Clark BJ, George WD, et al: Thrombocytopenia megalovirus. Blood 95:487-493, 2000.
with absent radii (TAR) syndrome: a new increased cellular 17. McAuley J, Boyer D, Patel D, et al: Early and longitudinal
radiosensitivity syndrome. Clin Oncol (R Coll Radiol) 7:56- evaluations of treated infants and children and untreated
58, 1995. historical patients with congenital toxoplasmosis: the
5. Hall JG: Thrombocytopenia and absent radius (TAR) syn- Chicago Collaborative Treatment Trial. Clin Infect Dis 18:38-
drome. J Med Genet 24:79-83, 1987. 72, 1994.
6. Freedman MH, Doyle JJ: Inherited bone marrow failure syn- 18. Sullivan EM, Burgess MA, Forrest JM: The epidemiology of
dromes. In Lilleyman JS, Hann IM, Branchette VS, editors: rubella and congenital rubella in Australia, 1992-1997.
Pediatric hematology, London, 1999, Churchill Livingstone, Commun Dis Intell 23:209-214, 1999.
pp 23-49. 19. Tookey PA, Peckham CS: Surveillance of congenital rubella
7. Lackner A, Basu M, Bierings M, et al: Haematopoietic stem in Great Britain, 1971-96. BMJ 318:769-770, 1999.
cell transplantation for amegakaryocytic thrombocytopenia. 20. Tovo PA, de-Martino M, Gabiano C, et al: Prognostic factors
Br J Haematol 109:773-775, 2000. and survival in children with perinatal HIV-1 infection. The
8. van den Oudenrijn SM, Bruin M, Folman CC, et al: Muta- Italian Register for HIV Infections in Children. Lancet
tions in the thrombopoietin receptor, Mlp, in children with 339:1249-1253, 1992.
congenital amegakaryocytic thrombocytopenia. Br J Haema- 21. Triplett DA, editor: Platelet function: laboratory evaluation and
tol 110:441-448, 2000. clinical application, Chicago, 1978, American Society of
9. Stormorken H, Hellum B, Egeland T, et al: X-linked throm- Clinical Pathologists.
bocytopenia and thrombocytopathia: attenuated Wiscott- 22. Brown BA: Hematology: principles and procedures, ed 6,
Aldrich syndrome: functional and morphological studies of Philadelphia, 1993, Lea & Febiger.
platelets and lymphocytes. Thromb Haemost 65:300-305, 23. Aster R: Drug-induced thrombocytopenia. In Michelson
1991. AD, editor: Platelets, San Diego, 2002, Academic Press,
10. Thorup OA: Leavell and Thorups fundamentals of clinical pp 593-606.
hematology, ed 5, Philadelphia, 1987, Saunders. 24. Vadhan-Raj S: Clinical experience with recombinant human
11. Villa AL, Notarangelo P, Maachi E, et al: X-linked thrombo- thrombopoietin in chemotherapy-induced thrombocytope-
cytopenia and Wiskott-Aldrich syndrome are allelic diseases nia. Semin Hematol 37:28-34, 2000.
with mutations in the WASP gene. Nat Genet 9:414-417, 25. Basser RL, Underhill C, Davis MD, et al: Enhancement of
1999. platelet recovery after myelosuppressive chemotherapy by
12. Mehaffey MG, Newton AL, Gandhi MJ, et al: X-linked recombinant human megakaryocyte growth and develop-
thrombocytopenia caused by a novel mutation of GATA-1. ment factor in patients with advanced cancer. J Clin Oncol
Blood 98:2681-2688, 2001. 18:2852-2861, 2000.
CHAPTER 43 Thrombocytopenia and Thrombocytosis 715
26. Koch MA, Volberding PA, Lagakos SW, et al: Toxic effects of 47. Jacobs P, Wood L, Novitzky N: Intravenous gammaglobulin
zidovudine in asymptomatic human immunodeficiency has no advantages over oral corticosteroids as primary
virusinfected individuals with CD4+ cell counts of 0.5 therapy for adults with immune thrombocytopenia: a pro-
109/L or less: detailed and updated results from Protocol spective randomized clinical trial. Am J Med 1:55-59, 1994.
019 of the AIDS Clinical Trials Group. Arch Intern Med 48. Reference deleted in pages.
152:2286-2292, 1992. 49. Kojouri K, Vesely SK, Terrell DR, et al: Splenectomy for adult
27. Silverstein MN, Tefferi A: Treatment of essential thrombocy- patients with idiopathic thrombocytopenic purpura. Blood
themia with anagrelide. Semin Hematol 36:23-25, 1999. 104:2623-2634, 2004.
28. Martin TG, Shuman MA: Interferon-induced thrombocyto- 50. Smith ME, Reid DM, Jones CE, et al: Binding of quinine-
penia: is it time for thrombopoietin? Hepatology 28:1430- and quinidine-dependent drug antibodies to platelets is
1432, 1998. mediated by the Fab domain of the immunoglobulin G and
29. Colvin BT: Thrombocytopenia. Clin Haematol 14:661-681, is not Fc dependent. J Clin Invest 79:912-917, 1987.
1985. 51. Pfueller SL, Bilsont RA, Logan D, et al: Heterogeneity
30. Sata M, Yano Y, Yoshiyama, et al: Mechanisms of thrombo- of drug-dependent platelet antigens and their antibodies
cytopenia induced by interferon therapy for chronic hepa- in quinine- and quinidine-induced thrombocytopenia:
titis B. J Gastroenterol 32:206-221, 1997. involvement of glycoproteins Ib, IIb, IIIa, and IX. Blood
31. Trannel TJ, Ahmed I, Goebert D: Occurrence of thrombocy- 11:190-198, 1988.
topenia in psychiatric patients taking valproate. Am J Psy- 52. Chong BH, Fawaz I, Chesterman CN, et al: Heparin-
chiatry 158:128-130, 2001. induced thrombocytopenia: mechanism of interaction of
32. Thompson AR, Harker LA: Manual of hemostasis and throm- the heparin-dependent antibody with platelets. Br J Haema-
bosis, ed 3, Philadelphia, 1983, FA Davis. tol 73:235-240, 1989.
33. Taghizadeh M: Megaloblastic anemias. In Harmening DM, 53. Amiral J, Bridley F, Dreyfus M, et al: Platelet factor 4 com-
editor: Clinical hematology and fundamentals of hemostasis, plexed to heparin is the target for antibodies generated in
ed 3, Philadelphia, 1997, FA Davis, pp 116-134. heparin-induced thrombocytopenia. Thromb Haemost 68:95
34. Corriveau DM, Fritsma GA, editors: Hemostasis and (letter), 1992.
thrombosis in the clinical laboratory, Philadelphia, 1988, JB 54. Brace LD, Fareed J, Tomeo J, et al: Biochemical and phar-
Lippincott. macological studies on the interaction of PK 10169 and its
35. Quick AJ: Hemorrhagic diseases and thrombosis, ed 2, subfractions with human platelets. Haemostasis 16:93-105,
Philadelphia, 1966, Lea & Febiger. 1986.
36. Davis GL: Quantitative and qualitative disorders of platelets. 55. Warkentin TE: Heparin-induced thrombocytopenia. In
In Stiene-Martin EA, Lotspeich-Steininger C, Koepke JA, Colman RW, Marder VJ, Clowes AW, et al, editors: Hemosta-
editors: Clinical hematology: principle, procedures, correlations, sis and thrombosis, ed 5, Philadelphia, 2006, Lippincott
ed 2, Philadelphia, 1998, Lippincott Williams & Wilkins, Williams & Wilkins, pp 1649-1663.
pp 717-734. 56. Rhodes GR, Dixon RH, Silver D: Heparin-induced throm-
37. Bussel J, Cines D: Immune thrombocytopenia, neonatal bocytopenia: eight cases with thrombotic-hemorrhagic
alloimmune thrombocytopenia, and post-transfusion complications. Am Surg 186:752-758, 1977.
purpura. In Hoffman R, Benz EJ, Shattil SJ, et al, editors: 57. Brace LD, Fareed J: An objective assessment of the interac-
Hematology: basic principles and practice, ed 3, New York, tion of heparin and its subfractions with human platelets.
2000, Churchill Livingstone, pp 2096-2114. Semin Thromb Hemost 11:190-198, 1985.
38. Kessler CM, Acs P, Mariani G: Acquired disorders of coagula- 58. Fratantoni JC, Pollet R, Gralnick HR: Heparin-induced
tion: the immune coagulopathies. In Colman RW, Hirsh J, thrombocytopenia: confirmation by in vitro methods. Blood
Marder VJ, et al, editors: Hemostasis and thrombosis: basic 45:395-401, 1975.
principles and clinical practice, ed 5, Philadelphia, 2006, 59. Sheridan D, Carter C, Kelton JG: A diagnostic test for
Lippincott Williams & Wilkins, pp 1061-1085. heparin-induced thrombocytopenia. Blood 67:27-30, 1986.
39. Woerner SJ, Abildgaard CF, French BN: Intracranial hemor- 60. Brace LD: Testing for heparin-induced thrombocytopenia
rhage in children with idiopathic thrombocytopenic by platelet aggregometry. Clin Lab Sci 5:80-81, 1992.
purpura. Pediatrics 67:453-460, 1981. 61. Collins JL, Aster RH, Moghaddam M, et al: Diagnostic
40. Dameshek W, Ebbe S, Greenberg L, et al: Recurrent acute testing for heparin-induced thrombocytopenia (HIT): an
idiopathic thrombocytopenic purpura. N Engl J Med enhanced platelet factor 4 complex enzyme linked immu-
269:646-653, 1963. nosorbent assay (PF4 ELISA). Blood 90(suppl 1):461,
41. Stasi R, Provan D: Management of immune thrombocyto- 1997.
penic purpura in adults. Mayo Clin Proc 79:504-522, 2004. 62. Gruel Y, Boizard B, Daffos F, et al: Determination of platelet
42. Kunicki TJ, Newman PJ: The molecular immunology of antigens and glycoproteins in the human fetus. Blood
human platelet proteins. Blood 80:1386-1404, 1992. 68:488-492, 1986.
43. Beer JH, Rabaglio M, Berchtold P, et al: Autoantibodies 63. Mueller-Eckhardt C, Kiefel V, Grubert A, et al: 348 cases of
against the platelet glycoproteins (GP) IIb/IIIa, Ia/IIa, and suspected neonatal alloimmune thrombocytopenia. Lancet
IV and partial deficiency in GPIV in a patient with a bleeding 1:363-366, 1989.
disorder and a defective platelet collagen interaction. Blood 64. McFarland JG, Aster RH, Bussel JB, et al: Prenatal diagnosis
82:820-829, 1993. of neonatal alloimmune thrombocytopenia using allele-
44. Olsson B, Andersson P, Jernas M, et al: T-cellmediated specific oligonucleotide probes. Blood 78:2276-2282, 1991.
cytotoxicity toward platelets in chronic idiopathic thrombo- 65. Levin AB, Berkowitz RL: Neonatal alloimmune thrombocy-
cytopenic purpura. Nat Med 9:1123-1124, 2003. topenia. Semin Perinatol 15(suppl 2):35-40, 1991.
45. George JN, Kojouri K: Immune thrombocytopenic purpura. 66. Kaplan C, Daffos F, Forestier F, et al: Management of alloim-
In Colman RW, Marder VJ, Clowes AW, et al, editors: Hemo- mune thrombocytopenia: antenatal diagnosis and in utero
stasis and thrombosis, ed 5, Philadelphia, 2006, Lippincott transfusion of maternal platelets. Blood 72:340-343, 1988.
Williams & Wilkins, pp 1085-1094. 67. Rayburn WF: Glucocorticoid therapy for rheumatic diseases:
46. Chong BH, Ho S-J: Autoimmune thrombocytopenia. maternal, fetal, and breast-feeding considerations. Am J
J Thromb Haemost 3:1763-1772, 2005. Reprod Immunol 28:138-140, 1992.
716 PART VIII Hemostasis and Thrombosis

68. Clerc JM: Neonatal thrombocytopenia. Clin Lab Sci 2:42-47, 90. Moake JL: Thrombotic thrombocytopenic purpura and
1989. hemolytic uremic syndrome. In Michelson AD, editor: Plate-
69. Mueller-Eckhardt C: Post-transfusion purpura. Br J Haematol lets, San Diego, 2002, Academic Press, pp 607-620.
64:419-424, 1986. 91. Bell WR, Braine HG, Ness PM, et al: Improved survival in
70. Vogelsang G, Kickler TS, Bell WR: Post-transfusion purpura: thrombotic thrombocytopenic purpurahemolytic uremic
a report of five patients and a review of the pathogenesis syndrome: clinical experience in 108 patients. N Engl J Med
and management. Am J Hematol 21:259-267, 1986. 325:398-403, 1991.
71. McLaughlin P, Talpaz M, Quesada JR, et al: Immune throm- 92. Rock GA, Shumak KH, Buskard NA, et al: Comparison of
bocytopenia following -interferon therapy in patients with plasma exchange with plasma infusion in the treatment of
cancer. JAMA 254:1353-1354, 1985. thrombotic thrombocytopenic purpura. N Engl J Med
72. Yoshida Y, Hirashima K, Asano S, et al: A phase II trial of 325:393-397, 1991.
recombinant human granulocyte colony-stimulating factor 93. Moake JL, Turner NA, Stathopoulos NA, et al: Involvement
in the myelodysplastic syndromes. Br J Haematol 78:378- of large plasma von Willebrand factor (VWF) multimers and
384, 1991. unusually large VWF forms derived from endothelial cells
73. Paciucci PA, Mandeli J, Oleksowicz L, et al: Thrombocyto- in shear-stress induced platelet aggregation. J Clin Invest
penia during immunotherapy with interleukin-2 by con- 78:1456-1461, 1986.
stant infusion. Am J Med 89:308-312, 1990. 94. Furlan M, Robles R, Solenthaler M, et al: Deficient activity
74. Fink K, Al-Mondhiry H: Idiopathic thrombocytopenic of von Willebrand factor-cleaving protease in chronic
purpura in lymphoma. Cancer 37:1999-2004, 1976. relapsing thrombotic thrombocytopenic purpura. Blood
75. Carey RW, McGinnis A, Jacobson BM, et al: Idiopathic 89:3097-3103, 1997.
thrombocytopenic purpura complicating chronic lympho- 95. Furlan M, Robles R, Lammle B: Partial purification and char-
cytic leukemia: management with sequential splenectomy acterization of a protease from human plasma cleaving von
and chemotherapy. Arch Intern Med 136:62-66, 1976. Willebrand factor to fragments produced by in vivo prote-
76. Miller MH, Urowitz MB, Gladman DD: The significance of olysis. Blood 87:4223-4234, 1996.
thrombocytopenia in systemic lupus erythematosus. Arthri- 96. Tsai HM: Physiologic cleaving of von Willebrand factor by
tis Rheum 26:1181-1186, 1983. a plasma protease is dependent on its conformation and
77. Kelton JG, Keystone J, Moore J, et al: Immune-mediated requires calcium ion. Blood 87:4235-4244, 1996.
thrombocytopenia of malaria. J Clin Invest 71:832-836, 97. Tsai HM, Lian ECY: Antibodies to von Willebrand factor
1983. cleaving protease in acute thrombotic thrombocytopenic
78. Barron WM: The syndrome of preeclampsia. Gastroenterol purpura. N Engl J Med 339:1585-1594, 1998.
Clin North Am 21:851-872, 1992. 98. Furlan M, Lammle B: von Willebrand factor in thrombotic
79. Schindler M, Gatt S, Isert P, et al: Thrombocytopenia and thrombocytopenic purpura. Thromb Haemost 82:592-600,
platelet function defects in pre-eclampsia: implications 1999.
for regional anesthesia. Anaesth Intensive Care 18:169-174, 99. Gerritsen HE, Turecek PL, Schwarz HP, et al: Assay of
1990. von Willebrand factor (VWF)cleaving protease based on
80. Gibson W, Hunter D, Neame PB, et al: Thrombocytopenia decreased collagen binding affinity of degraded VWF: a tool
in preeclampsia and eclampsia. Semin Thromb Hemost 8:234- for the diagnosis of thrombotic thrombocytopenic purpura
247, 1982. (TTP). Thromb Haemost 82:1386-1389, 1999.
81. Martin JN Jr, Blake PG, Perry KG Jr, et al: The natural history 100. Moake JL, McPherson PD: Abnormalities of von Willebrand
of HELLP syndrome: patterns of disease progression and factor multimers in thrombotic thrombocytopenic purpura
regression. Am J Obstet Gynecol 164:1500-1509, 1991. and the hemolytic uremic syndrome. Am J Med 87: 3-9,
82. Green D: Diagnosis and management of bleeding disorders. 1989.
Compr Ther 14:31-36, 1988. 101. Byrnes JJ, Moake JL, Klug P, et al: Effectiveness of the cryo-
83. Trofatter KF Jr, Howell ML, Greenberg CS, et al: Use of the supernatant fraction of plasma in the treatment of refractory
fibrin D-dimer in screening for coagulation abnormalities thrombotic thrombocytopenic purpura. Am J Hematol
in preeclampsia. Obstet Gynecol 73:435-440, 1989. 34:169-174, 1990.
84. Burrows RF, Hunter DJS, Andrew M, et al: A prospective 102. Kwaan HC, Soff GA: Management of thrombotic thrombo-
study investigating the mechanism of thrombocytopenia in cytopenic purpura and hemolytic uremic syndrome. Semin
preeclampsia. Obstet Gynecol 70:334-338, 1987. Hematol 34:159-166, 1997.
85. Benigni A, Gregorini G, Frusca T, et al: Effect of low-dose 103. Karmali MA: The association of verotoxins and the classical
aspirin on fetal and maternal generation of thromboxane hemolytic uremic syndrome. In Kaplan BS, Trompeter RS,
by platelets in women at risk for pregnancy-induced hyper- Moake JL, editors: Hemolytic-uremic syndrome and thrombotic
tension. N Engl J Med 321:357-362, 1989. thrombocytopenic purpura, New York, 1992, Marcel Dekker,
86. Schiff E, Peleg E, Goldenberg M, et al: The use of aspirin to pp 199-212.
prevent pregnancy-induced hypertension and lower the 104. Obrig TG: Pathogenesis of Shiga toxin (verotoxin)induced
ratio of thromboxane A2 to prostacyclin in relatively high endothelial cell injury. In Kaplan BS, Trompeter RS, Moake
risk pregnancies. N Engl J Med 321:351-356, 1989. JL, editors: Hemolytic-uremic syndrome and thrombotic throm-
87. Ezra Y, Rose M, Eldor A: Therapy and prevention of throm- bocytopenic purpura, New York, 1992, Marcel Dekker,
botic thrombocytopenic purpura during pregnancy: a clini- pp 405-419.
cal study of 16 pregnancies. Am J Hematol 51:1-6, 1996. 105. Charba D, Moake JL, Harris MA, et al: Abnormalities of
88. Dashe JS, Ramin SM, Cunningham FG: The long-term von Willebrand factor multimers in drug-associated
consequences of thrombotic microangiopathy (thrombotic thrombotic microangiopathies. Am J Hematol 42:268-277,
thrombocytopenic purpura and hemolytic uremic syn- 1993.
drome) in pregnancy. Obstet Gynecol 91:662-668, 1998. 106. Santhosh-Kumar CR, Yohannon MD, Higgy KE, et al:
89. Moschcowitz E: An acute febrile pleiochromic anemia with Thrombocytosis in adults: analysis of 777 patients. J Intern
hyaline thrombosis of the terminal arterioles and capillar- Med 229:493-495, 1991.
ies: a hitherto undescribed disease. Arch Intern Med 36:89- 107. Mitus AJ, Schafer AI: Thrombocytosis and thrombocythe-
93, 1925. mia. Hematol Oncol Clin North Am 4:157-178, 1990.
CHAPTER 43 Thrombocytopenia and Thrombocytosis 717
108. Michiels JJ, Ten Cate FJ: Erythromelalgia in thrombocythe- 113. Lazzarino M, Vitale A, Morra E, et al: Interferon alpha-2b as
mia of various myeloproliferative disorders. Am J Hematol treatment for Philadelphia-negative myeloproliferative dis-
39:131-136, 1992. orders with excessive thrombocytosis. Br J Haematol 72:173-
109. Hoffman R: Primary thrombocythemia. In Hoffman R, Benz 177, 1989.
EJ, Shattil SJ, et al, editors: Hematology: basic principles 114. Giles FJ, Singer CRJ, Gray AG, et al: Alpha interferon for
and practice, ed 3, New York, 2000, Churchill Livingstone, essential thrombocythemia. Lancet 2:70-72, 1988.
pp 1188-1204. 115. Petitt RM, Silverstein MN, Petrone ME: Anagrelide for
110. Schafer AI: Bleeding and thrombosis in the myeloprolifera- control of thrombocythemia in polycythemia and other
tive disorders. Blood 64:1-12, 1984. myeloproliferative disorders. Semin Hematol 34:51-54,
111. Panlilio AL, Reiss RF: Therapeutic plateletpheresis in throm- 1997.
bocythemia. Transfusion 19:147-152, 1979. 116. Tefferi A, Silverstein MN, Petitt RM, et al: Anagrelide as a
112. Middelhoff G, Boll I: A long term trial of interferon alpha- new platelet-lowering agent in essential thrombocythemia:
therapy in essential thrombocythemia. Ann Hematol 64:207- mechanism of action, efficacy, toxicity, current indications.
209, 1992. Semin Thromb Hemost 23:379-383, 1997.
44 QualitativeandDisorders of Platelets
Vasculature
Larry D. Brace

OUTLINE OBJECTIVES
Qualitative Platelet After completion of this chapter, the reader will be able to:
Disorders
Disorders of Adhesion 1. Describe the effect of aspirin on the cyclooxygenase 7. Distinguish among the following types of inherited
Receptors pathway. platelet disorders: membrane receptor abnormality,
Disorders of Platelet 2. Describe the defect in each of the following heredi- secretion disorder, and storage pool deficiency.
Secretion (Release tary disorders: storage pool disease, gray platelet 8. For each of the inherited platelet disorders listed,
Reactions) syndrome, Glanzmann thrombasthenia, and Bernard- name useful laboratory tests and recognize diagnos-
Inherited Disorders of Soulier syndrome (BSS). tic results.
Other Receptors and 3. Discuss the mechanisms of action of antiplatelet 9. Identify the most common type of hereditary platelet
Signaling Pathways drugs. defect.
Acquired Defects of 4. Explain the effects of paraproteins on platelet 10. Discuss the mechanism of the platelet defects asso-
Platelet Function
function. ciated with myeloproliferative diseases, uremia, and
Vascular Disorders
5. Compare and contrast Glanzmann thrombasthenia liver disease.
Hereditary Vascular
Disorders and BSS. 11. Recognize conditions associated with acquired vas-
Acquired Vascular 6. Recognize the clinical presentation of patients with cular disorders.
Disorders dysfunctional platelets.

CASE STUDY
After studying the material in this chapter, the reader should be able to respond to the following case study:

A 19-year-old woman with a chief complaint of easy bruising, epinephrine induced only primary aggregation, and collagen-
occasional mild nosebleeds, and heavy periods was examined induced aggregation was decreased, although a near-normal
by her physician. At the time of her examination, she had a aggregation response could be obtained with a high collagen
few small bruises on her arms and legs, but no other prob- concentration. Spontaneous aggregation was not observed.
lems. Initial laboratory data revealed a prolonged bleeding 1. What are three possible explanations for the test results
time of 10 minutes, a normal prothrombin time, and a so far?
normal partial thromboplastin time. A CBC, including plate- 2. Given the bleeding history in her family, which of the
let count and morphology, yielded normal results. three explanations seems most likely?
A detailed history revealed that the patients bleeding prob- A quantitative test for adenosine triphosphate (ATP)
lems occurred most frequently after aspirin ingestion. Her release was performed using the firefly luciferin-luciferase
mother and one of her brothers also had some of the same bioluminescence assay. The result of this test showed a
symptoms. Blood was drawn for platelet function studies, marked decrease in the amount of ATP released when
and platelet aggregation tests were performed. Aggregation platelets were stimulated with thrombin.
induced by ristocetin and adenosine diphosphate was near 3. Based on the ATP test results, what is the likely cause of
normal, arachidonic acidinduced aggregation was absent, the patients bleeding symptoms?

718
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 719

C
linical manifestations of bleeding disorders can be
divided into two broad, rather poorly defined groups: BOX 44-1 Qualitative Abnormalities: Changes in
(1) superficial bleeding (e.g., petechiae, epistaxis, or Platelet Function (Thrombocytopathy)
gingival bleeding), usually associated with a platelet defect or Disorders of platelet adhesion
vascular disorder; and (2) deep tissue bleeding (e.g., hemato- Bernard-Soulier syndrome (giant platelet syndrome)
mas or hemarthrosis), usually associated with plasma clotting von Willebrand disease
factor deficiencies.1 This chapter describes platelet and vascular Acquired defects of platelet adhesion
disorders. Bleeding problems resulting from defects in the Myeloproliferative and lymphoproliferative disorders and
coagulation mechanism are described in Chapter 41. dysproteinemias
Antiplatelet antibodies
Cardiopulmonary bypass surgery
QUALITATIVE PLATELET DISORDERS Chronic liver disease
Excessive bruising, superficial (mucocutaneous) bleeding, and Drug-induced membrane modification
a prolonged bleeding time in a patient whose platelet count Disorders of primary aggregation
is normal suggest an acquired or a congenital disorder of plate- Glanzmann thrombasthenia (congenital)
let function. Congenital disorders have been described that Hereditary afibrinogenemia
result from abnormalities of each of the major phases of plate- Acquired
let function: adhesion, aggregation, and secretion. Rapid pro Acquired von Willebrand disease
gress in this field began in the 1960s, mostly as a result of the Uremia
development of instruments and test methods for measuring Disorders of platelet secretion (release reactions)
platelet function.2 Qualitative disorders are summarized in Storage pool diseases
Box 44-1. Dense granule deficiencies
Hermansky-Pudlak syndrome
Disorders of Adhesion Receptors Wiskott-Aldrich syndrome
Bernard-Soulier (Giant Platelet) Syndrome Thrombocytopeniaabsent radius (TAR) syndrome
Bernard-Soulier syndrome (BSS) is a rare syndrome that is Chdiak-Higashi syndrome
usually manifested in infancy or childhood with hemorrhage -Granule deficiencies
characteristic of defective platelet function: ecchymoses, epi- Gray platelet syndrome
staxis, and gingival bleeding. Hemarthroses and expanding Thromboxane pathway disorders: aspirin-like defects
hematomas are only rarely seen. BSS is inherited as an autoso- Hereditary aspirin-like defects
mal recessive disorder in which the glycoprotein Ib/IX/V (GP Cyclooxygenase or thromboxane synthetase deficiency
Ib/IX/V) complex is missing from the platelet surface or exhib- Drug inhibition of the prostaglandin pathways
its abnormal function. Heterozygotes who have about 50% Drug inhibition of platelet phosphodiesterase activity
of normal levels of GP Ib, GP V, and GP IX have normal or Changes in membrane phospholipid distribution
near-normal platelet function. Homozygotes have a moderate Scott syndrome
to severe bleeding disorder characterized by a prolonged bleed- Stormorken syndrome
ing time, enlarged platelets, thrombocytopenia, and usually Hyperactive prethrombotic platelets
decreased platelet survival. Platelet counts generally range from Data from Thompson AR, Harker LA: Manual of hemostasis and thrombosis,
40,000/mcL to near normal.3 On peripheral blood films, plate- ed 3, Philadelphia, 1983, FA Davis; and Colvin BT: Thrombocytopenia, Clin
lets typically are 5 to 8m in diameter, but can be 20m Haematol 14:661-681, 1985.
(Figure 44-1). Viewed by electron microscopy, BSS platelets
contain a larger number of cytoplasmic vacuoles and mem-
brane complexes, and these observations extend to megakaryo- essential to normal function because it contains binding sites
cytes, in which the appearance of the demarcation membrane for von Willebrand factor (VWF) and thrombin. Defects in the
system is irregular.2,4-7 GP Ib and GP IX genes also are known to result in BSS,
Four glycoproteins are required to form the GP Ib/IX/V however.8-10
complex: GP Ib, GP Ib, GP IX, and GP V. In the complex, BSS platelets have normal aggregation responses to adeno
these proteins are present in the ratio of 2:2:2:1. The gene for sine diphosphate (ADP), epinephrine, collagen, and arachi-
GP Ib is located on chromosome 17, the gene for GP Ib is donic acid but do not respond to ristocetin and have
located on chromosome 22, and the genes for GP IX and GP diminished response to thrombin.2,4,6 The lack of response to
V are located on chromosome 3. For surface expression of the ristocetin is due to the lack of GP Ib/IX/V complexes and the
GP Ib/IX complex, it seems that synthesis of three proteins, GP inability of BSS platelets to bind VWF. Lack of binding to VWF
Ib, GP Ib, and GP IX, is required. Only GP V can be expressed also accounts for the inability of platelets to adhere to exposed
alone in significant quantities on the surface of platelets, but subendothelium and the resultant bleeding characteristic of
the expression seems to be enhanced if the rest of the complex this disorder. This defect in adhesion shows the importance
is present. The most frequent forms of BSS involve defects in of initial platelet attachment in primary hemostasis. In many
GP Ib synthesis or expression. The presence of GP Ib is respects, this disorder resembles the defect seen in von
720 PART VIII Hemostasis and Thrombosis

bleeding. Hemorrhagic manifestations include petechiae, pur


pura, menorrhagia, gastrointestinal bleeding, and hematuria.
There are wide variations in the clinical symptoms. Some
patients may have minimal symptoms, whereas others may
have frequent and serious hemorrhagic complications. The
severity of the bleeding episodes seems to decrease with age.14,15
The biochemical lesion responsible for the disorder is a
deficiency or abnormality of the platelet membrane GP IIb/IIIa
(IIb/3) complex, a membrane receptor capable of binding
fibrinogen, VWF, fibronectin, and other adhesive ligands. Typi-
cally, the platelets of homozygous individuals lack surface-
expressed GP IIb/IIIa, whereas the GP IIb/IIIa content of the
platelets from heterozygotes has been found to be 50% to
60% of normal.4,16 Binding of fibrinogen to the GP IIb/IIIa
complex mediates normal platelet aggregation responses.
Failure of such binding results in a profound defect in hemo-
static plug formation and the serious bleeding characteristic of
Figure 44-1 Giant platelets in Bernard-Soulier syndrome (peripheral thrombasthenia.2,4,6,17-19
blood, 1000). (Modified from Carr JH, Rodak BF: Clinical hematology atlas, More than 70 mutations are known to give rise to Glan-
ed 3, Philadelphia, 2009, Saunders.) zmann thrombasthenia.20-22 The IIb and 3 genes are present
on chromosome 17, and genetic defects are distributed widely
over the two genes. IIb is synthesized in megakaryocytes as
Willebrand disease (VWD). In contrast to VWD, however, this pro-IIb, which complexes 3 in the endoplasmic reticulum.
abnormality cannot be corrected by the addition of normal The complex is transported to the Golgi body, where IIb is
plasma or cryoprecipitate, which is consistent with a defect that cleaved to heavy and light chains to form the complete complex.
resides in the platelets. Uncomplexed IIb and 3 are not processed in the Golgi body.
Several unusual variants of BSS have been described in As with the GP Ib/IX/V complex, it is necessary for both pro-
which all or most of the GP Ib/IX/V complex is present, but teins of the GP IIb/IIIa complex to be produced and assembled
the mutations affect binding domains, affect interactions into a complex for the complex to be expressed on the platelet
between elements of the complex, or result in truncation of a surface. Gene defects that lead to the absence of production of
specific protein in the complex. In these cases, the complex fails either protein lead to absence of the complex on the platelet
to bind VWF or does so poorly.9 surface. Defects that interfere with or prevent complex forma-
Platelet-type VWD (pseudo-VWD) is associated with a gain tion or affect complex stability have the same effect.
of function and spontaneous binding of plasma VWF. GP Ib Numerous variants of Glanzmann thrombasthenia have
mutations that give rise to platelet-type VWD are 233Gly Val been described in which the GP IIb/IIIa complex is qualita-
or Ser and 239MetVal.11,12 Loss of residues 421 to 429 in GP tively abnormal. IIb and 3 are produced, form a complex, and
Ib also has been reported to result in platelet-type VWD.13 are processed normally. One or more functions of the complex
There is no specific treatment for BSS. Platelet transfusions (e.g., fibrinogen binding or signal transduction) are abnormal,
are the therapy of choice, but patients invariably develop allo- however. Bleeding in these patients ranges from mild to severe.
antibodies, so that further platelet transfusion is not possible. One component of the IIb/3 integrin, 3, is a component
BSS patients tend to do better if apheresis platelets are used for of the vitronectin receptor, V/3, found on endothelial cells,
transfusion, because this tends to limit the number of donors osteoclasts, fibroblasts, monocytes, and activated B lympho-
to which the patient is exposed, and the rate of alloimmuniza- cytes, where it acts as a receptor for a variety of adhesive protein
tion tends to be lower.4,6 Other treatments that have been used ligands. Patients who have 3 gene defects that result in the
include desmopressin acetate (DDAVP) and, more recently, absence of IIb/3 integrin also lack the vitronectin receptor.
recombinant factor VIIa. These patients do not seem to have a more severe form of
Glanzmann thrombasthenia.23,24 The vitronectin receptor is
Glanzmann Thrombasthenia thought to play a role in vascularization, but so far no evidence
Glanzmann thrombasthenia originally was described as a for abnormal blood vessel development has been documented
bleeding disorder associated with abnormal in vitro clot in individuals lacking the vitronectin receptor. It also is unclear
retraction and a normal platelet count. It is inherited as an whether platelet vitronectin receptors play any significant role
autosomal recessive disorder and is seen most frequently in in platelet functional processes.20
populations with a high degree of consanguinity. Heterozy- The typical laboratory features of Glanzmann thrombasthe-
gotes are clinically normal, whereas homozygotes have serious nia are a markedly prolonged bleeding time, a normal platelet
bleeding problems. This rare disorder manifests itself clinically count and normal platelet morphology, poor in vitro clot
in the neonatal period or infancy, occasionally with bleeding retraction, and a lack of platelet aggregation in response
after circumcision and frequently with epistaxis and gingival to all platelet-activating agents (including ADP, collagen,
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 721
thrombin, and epinephrine).2,4,6,18 If the stimulating agent is enhance thrombus formation at the site of a lesion by stimulat-
strong enough (e.g., thrombin), the platelets undergo the ing tissue factorindependent thrombus generation and fibrin
release/secretion reaction, even in the absence of aggregation. formation.34
Ristocetin-induced binding of VWF to platelets and the result- Rarely, a thrombasthenia-like state can be acquired. Such
ing platelet agglutination are normal. The results of the com- conditions include development of autoantibodies against GP
plete blood count (CBC) are usually normal, unless there is IIb/IIIa, multiple myeloma in which the paraprotein is directed
another underlying disorder or the patient has had a recent against GP IIIa, and afibrinogenemia. A thrombasthenia-like
hemorrhagic episode. Tests for platelet procoagulant activity, state also can be induced in individuals with otherwise normal
previously called the platelet factor 3 test, usually show dimin- platelet function by the therapeutic antiplatelet drugs ticlopi-
ished activity.2,16,25 There seem to be several reasons for this. dine and clopidogrel.15,18
When normal platelets are activated, procoagulant microvesi-
cles are shed from the platelet surface, and coagulation factors Disorders of Platelet Secretion
assemble on the microvesicle surfaces during activation of the (Release Reactions)
coagulation cascade. In Glanzmann thrombasthenia, markedly Of the hereditary platelet function defects, disorders involving
fewer microvesicles are produced. Second, prothrombin binds storage pool defects and the release reaction are the most
directly to GP IIb/IIIa. Because this complex is missing in common. The clinical features of this group of disorders are
Glanzmann thrombasthenia, significantly less thrombin is mucocutaneous hemorrhage and hematuria, epistaxis, and
generated in response to tissue factor. Finally, Glanzmann easy and spontaneous bruising. Petechiae are less common
thrombasthenia platelets are not as activated by thrombin as than in other qualitative platelet disorders. Hemorrhage is
are normal platelets.26-29 rarely severe but may be exacerbated by ingestion of aspirin or
A subdivision of Glanzmann thrombasthenia into cases other antiplatelet agents. In most of these disorders, the platelet
with absent (type 1) and subnormal (type 2) in vitro clot count is normal, and the bleeding time is usually, although not
retraction has been proposed. In general, individuals with type always, prolonged. Platelet aggregation abnormalities are
2 disease have more residual GP IIb/IIIa complexes (10% to usually seen but vary depending on the disorder.2,4,35,36
20% of normal) than those with type 1 disease (0% to 5% of
normal), although there is considerable variability within each Storage Pool Diseases
subdivision.30,31 Dense Granule Deficiencies. The inheritance of -
Thrombasthenia is one of the few forms of platelet dysfunc- granule (dense granule) deficiency does not follow a single
tion in which hemorrhage is severe and disabling. Bleeding of mode, and it is likely that a variety of genetic abnormalities
all types, including epistaxis, ecchymosis, hemarthrosis, subcu- lead to the development of this disorder. Dense granule defi-
taneous hematoma, menorrhagia, and gastrointestinal and ciencies can be subdivided into deficiency states associated
urinary tract hemorrhage, has been reported. Treatment of with albinism and those in otherwise normal individuals (non-
bleeding episodes in patients with Glanzmann thrombasthenia albinos). In the platelets of nonalbinos, there is evidence for
requires the transfusion of normal platelets. In Glanzmann the presence of -granule membranes in normal to near-normal
thrombasthenia, the defective platelets may interfere with the numbers, which suggests that the disorder arises from an
normal transfused platelets, and it may be necessary to infuse inability to package the -granule contents.37,38 Serotonin accu-
more donor platelets than expected to control bleeding. As in mulates in normal -granules by an active uptake mechanism
BSS or any situation in which repeated transfusions are required, in which plasma serotonin is transported by a specific carrier-
patients with Glanzmann thrombasthenia may become alloim- mediated system across the plasma membrane into the
munized. Strategies to reduce alloimmunization include use of cytoplasm, and a second carrier-mediated system in the
single-donor platelet apheresis products, HLAmatched donor -granule membrane transports serotonin from the cytoplasm
platelets, or ABO-matched donor platelets.32 to the interior of the -granules.39 These transport mechanisms
A variety of treatments have been used successfully to are used in the serotonin release assay employed to detect
control or prevent bleeding, alone or in combination with heparin-dependent antiplatelet antibodies (see Chapter 43). In
platelet transfusion. To a large extent, the site of hemorrhage addition to serotonin transport mechanisms, a nucleotide
determines the therapeutic approach used. Hormonal therapy transporter MRP4 (ABCC4) that is highly expressed in platelets
(norethindrone acetate) has been used to control menorrhagia. and -granules has been identified. It would be expected that
If the patient is treated with oral contraceptives, excessive mutations in the gene for this transporter could affect nucleo-
bleeding should be reduced. Menorrhagia at the onset of tide accumulation in -granules.40
menses is uniformly severe and can be life-threatening, which As an isolated abnormality, -granule deficiency does not
has led some to suggest that birth control pills be started before typically result in a serious hemorrhagic problem. Bleeding is
menarche. Also, antifibrinolytic therapy (aminocaproic acid or usually mild and most often is limited to easy bruisability.
tranexamic acid) can be used to control gingival hemorrhage Dense granule deficiency affects the results of platelet aggrega-
or excessive bleeding after tooth extraction.31 Recombinant tion tests. -Granules are intracellular storage sites for ADP,
factor VIIa has proved useful to treat severe bleeding in patients adenosine triphosphate, calcium, pyrophosphate, and sero-
with isoantibodies to IIb/3 and in patients undergoing inva- tonin. The contents of these granules are extruded when plate-
sive procedures.33 Recombinant factor VIIa is thought to let secretion is induced, and secreted ADP plays a major role
722 PART VIII Hemostasis and Thrombosis

in the propagation of platelet activation, recruitment, and owing to immune deficiency, and life-threatening thrombocy-
aggregation and growth of the hemostatic plug. In patients topenia. Individuals with this disorder lack the ability to make
with -granule deficiency, addition of arachidonic acid to antipolysaccharide antibodies, which results in a propensity for
platelet-rich plasma fails to induce an aggregation response. pneumococcal sepsis. Bleeding episodes are typically moderate
Epinephrine and low-dose ADP induce a primary wave of to severe. A milder form without immune deficiency is known
aggregation, but a secondary wave is missing. Responses to low as hereditary X-linked thrombocytopenia (see Chapter 43). In
concentrations of collagen are decreased to absent, but a high Wiskott-Aldrich syndrome, a combination of ineffective throm-
concentration of collagen may induce a near-normal aggrega- bocytopoiesis and increased platelet sequestration and destruc-
tion response.36,39 This aggregation pattern is caused by the lack tion accounts for the thrombocytopenia. As with all X-linked
of ADP secretion and is almost identical to the pattern observed recessive disorders, it is found primarily in males.6,9,16,25,44 The
in patients taking aspirin. gene that is mutated in Wiskott-Aldrich syndrome codes for
In addition to occurring as an isolated problem, -granule a 502-amino-acid protein, WASP, that is found exclusively
deficiency is found in association with several disorders. in hematopoietic cells. WASP is primarily involved in signal
Hermansky-Pudlak syndrome is an autosomal recessive disor- transduction.
der characterized by tyrosinase-positive oculocutaneous albi- Wiskott-Aldrich platelets are also structurally abnormal. The
nism, defective lysosomal function in a variety of cell types, number of -granules is decreased, and the platelets are small,
ceroid-like deposition in the cells of the reticuloendothelial a feature of diagnostic importance. Other than in Wiskott-
system, and a profound platelet -granule deficiency.41 Several Aldrich syndrome, such small platelets are seen only in
of the mutations responsible for Hermansky-Pudlak syndrome TORCH (Toxoplasma, other agents, rubella virus, cytomegalovi-
have been mapped to chromosome 19. Mutations in at least rus, herpesvirus) infections. Diminished levels of stored
seven genes individually can give rise to Hermansky-Pudlak adenine nucleotides are reflected in the lack of -granules
syndrome. These genes encode for proteins that are involved observed on transmission electron micrographs. The platelet
in intracellular vesicular trafficking and are active in the bio- aggregation pattern in Wiskott-Aldrich syndrome is typical of
genesis of organelles.42 Whereas the bleeding associated with a storage pool deficiency. The platelets show a decreased aggre-
most -granule deficiencies is rarely severe, Hermansky-Pudlak gation response to ADP, collagen, and epinephrine and lack a
syndrome seems to be an exception. Although most bleeding secondary wave of aggregation in response to these agonists.
episodes in Hermansky-Pudlak syndrome are not severe, lethal The response to thrombin is normal, however.6,25 The most
hemorrhage has been reported, and in one series hemorrhage effective treatment for the thrombocytopenia seems to be sple-
accounted for 16% of deaths in patients with Hermansky- nectomy, which would be consistent with peripheral destruc-
Pudlak syndrome. A unique morphologic abnormality has tion of platelets. Platelet transfusions may be needed to treat
been described in the platelets of four families with Hermansky- hemorrhagic episodes. Bone marrow transplantation also has
Pudlak syndrome. This abnormality consists of marked dila- been attempted with some success.25,45
tion and tortuosity of the surface-connecting tubular system Thrombocytopenia with absent radii (TAR) syndrome (see
(the so-called Swiss cheese platelet).2,6,25,43 Chapter 43) is a rare autosomal recessive disorder characterized
Chdiak-Higashi syndrome is a rare autosomal recessive by the congenital absence of the radial bones (the most pro-
disorder characterized by partial oculocutaneous albinism, fre- nounced skeletal abnormality), numerous cardiac and other
quent pyogenic bacterial infections, giant lysosomal granules skeletal abnormalities, and thrombocytopenia (90% of cases).
in cells of hematologic (see Figure 28-5) and nonhematologic It is mentioned here because the platelets have structural
origin, platelet -granule deficiency, and hemorrhage. The defects in -granules with corresponding abnormal aggrega-
Chdiak-Higashi syndrome protein gene is located on chromo- tion responses. Marrow megakaryocytes may be decreased in
some 13, and a series of nonsense and frameshift mutations number, immature, or normal.6,46
all result in a truncated Chdiak-Higashi syndrome protein that
gives rise to a disorder of generalized cellular dysfunction -Granule Deficiency: Gray Platelet Syndrome. The
involving fusion of cytoplasmic granules. The disorder pro- -granules are the storage site for proteins produced by the
gresses to an accelerated phase in 85% of patients with Chdiak- megakaryocyte (e.g., platelet-derived growth factor, thrombos-
Higashi syndrome and is marked by lymphocytic proliferation pondin, and platelet factor 4) or present in plasma and taken
in the liver, spleen, and marrow and macrophage accumulation up by platelets and transported to -granules for storage (e.g.,
in tissues. During this stage, the pancytopenia worsens, which albumin, immunoglobulin G [IgG], and fibrinogen). There are
leads to hemorrhage and ever-increasing susceptibility to infec- 50 to 80 -granules per platelet, which are primarily respon-
tion; the result is death at an early age. Initially, the bleeding sible for the granular appearance of platelets on stained blood
time is increased because of -granule deficiency and conse- films. Gray platelet syndrome, a rare disorder first described in
quent defective platelet function. During the accelerated phase, 1971, is characterized by the specific absence of morpholo
however, the thrombocytopenia also contributes to the pro- gically recognizable -granules in platelets. The disorder is
longed bleeding time. Bleeding episodes vary from mild to inherited in an autosomal recessive fashion. Clinically, gray
moderate but worsen as the platelet count decreases.6,25 platelet syndrome is characterized by lifelong mild bleeding
Wiskott-Aldrich syndrome is an X-linked recessive disease tendencies, prolonged bleeding time, moderate thrombocyto-
characterized by the triad of severe eczema, recurrent infections penia, fibrosis of the marrow, and large platelets whose gray
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 723
appearance on a Wright-stained blood film is the source of the arachidonic acid depends on the degree of inhibition. Throm-
name of this disorder.2,4,6,47 boxane A2 is required for storage granule secretion and maximal
In electron photomicrographs of platelets and megakaryo- platelet aggregation in response to epinephrine, ADP, and low
cytes, the platelets appear to have virtually no -granules, concentrations of collagen.6,14,25,51
although they do contain vacuoles and small -granule precur- Hereditary absence or abnormalities of the components of
sors that stain positive for VWF and fibrinogen. The other types the thromboxane pathway are usually termed aspirin-like defects
of granules are present in normal numbers. The membranes of because the clinical and laboratory manifestations resemble
the vacuoles and the -granule precursors have P-selectin those that follow aspirin ingestion. Platelet aggregation
(CD62) and GP IIb/IIIa, and these proteins can be translocated responses are similar to those in storage pool disorders (see
to the cell membrane on stimulation with thrombin. This indi- earlier). Unlike in storage pool disorders, however, ultrastruc-
cates that these structures are -granules that cannot store the ture and granular contents are normal. Deficiencies of the
typical -granule proteins. This may provide an explanation for enzymes cyclooxygenase and thromboxane synthase are well
the observation that, in gray platelet syndrome, the plasma documented, and dysfunction or deficiency of thromboxane
levels of platelet factor 4 and -thromboglobulin are increased. receptors is known.38
Most patients develop early-onset myelofibrosis, which can be
attributed to the inability of megakaryocytes to store newly Inherited Disorders of Other Receptors and
synthesized platelet-derived growth factors.38 Signaling Pathways
Treatment of severe bleeding episodes may require platelet The 21 (GP Ia/IIa) integrin is one of the collagen receptors
transfusions. Few other treatments are available for these in the platelet membrane. A deficiency of this receptor has been
patients. Cryoprecipitate has been used to control bleeding. reported in a patient who lacked an aggregation response to
Desmopressin acetate was found to shorten the bleeding time collagen, whose platelets did not adhere to collagen, and who
and has been used as successful prophylaxis in a dental extrac- had a lifelong mild bleeding disorder.52 A deficiency in another
tion procedure. Some authors believe that desmopressin acetate collagen receptor, GP VI, also has been reported in patients
should be the initial therapy of choice.2,4,38,48,49 with mild bleeding. The platelets of these patients failed to
aggregate in response to collagen, and adhesion to collagen
Other Storage Pool Diseases. A rare disorder in which also was impaired.53 A family with gray platelet syndrome and
both - and -granules are deficient is known as - storage pool defective collagen adhesion has been described. Affected
deficiency. It seems to be inherited as an autosomal dominant members of the family have a severe deficiency of GP VI.54
characteristic. In these patients, other membrane abnormalities Platelets seem to contain at least three receptors for ADP.
also have been described.9 P2X1 is linked to an ion channel that facilitates calcium ion
Quebec platelet disorder is an autosomal dominant bleed- influx. P2Y1 and P2Y12 (P2TAC) are members of the seven-
ing disorder that results from a deficiency of multimerin (a transmembrane domain (STD) family of G proteinlinked
multimeric protein that is stored complexed with factor V in receptors. P2Y1 is thought to mediate calcium mobilization
-granules) and shows protease-related degradation of many and shape change in response to ADP. Pathology of the P2Y1
-granule proteins, even though -granule structure is main- receptor has not yet been reported. P2Y12 is thought to
tained. Thrombocytopenia may be present, although it is not be responsible for macroscopic platelet aggregation and is
a consistent feature.9 coupled to adenylate cyclase through a G-inhibitory (Gi)
protein complex.55 Some patients have been reported to have
Thromboxane Pathway Disorders: decreased platelet aggregation in response to ADP but normal
Aspirin-like Effects platelet shape change and calcium mobilization. These patients
Platelet secretion requires the activation of several biochemical have an inherited deficiency of the P2Y12 receptor.56-58 Bleeding
pathways. One such pathway is the one leading to thrombox- problems seem to be relatively mild in these patients, but the
ane formation. A series of phospholipases catalyze the release only treatment for severe bleeding is platelet transfusion.
of arachidonic acid and several other compounds from mem- Congenital defects of the 2-adrenergic (epinephrine) recep-
brane phospholipids. Arachidonic acid is converted to inter tor associated with decreased platelet activation and aggrega-
mediate prostaglandins by cyclooxygenase and to thromboxane tion in response to epinephrine are known. The receptors that
A2 by thromboxane synthase. Thromboxane A2 and other sub- mediate aggregation in response to epinephrine, ADP, and col-
stances generated during platelet activation cause mobilization lagen are STD receptors, as are the protease-activated receptors
of ionic calcium from internal stores into the cytoplasm, occu- (PARs) for thrombin. So far, defects in the PAR receptors have
pancy of several activation receptors, and initiation of a cascade not been described.9
of events resulting in secretion and aggregation of platelets (see A group of intracellular defects that affect platelet function
Chapter 13).50 includes defects in which all elements of the thromboxane
Several acquired or congenital disorders of platelet secretion pathway are normal, but insufficient calcium is released from
can be traced to structural and functional modifications of the dense tubular system, and the cytoplasmic concentration
arachidonic acid pathway enzymes. Inhibition of cyclooxygen- of ionic calcium in the cytoplasm never reaches levels high
ase occurs on ingestion of drugs such as aspirin and ibuprofen. enough to support secretion. This group of disorders is often
As a result, the amount of thromboxane A2 produced from referred to as calcium mobilization defects. These represent a
724 PART VIII Hemostasis and Thrombosis

heterogeneous group of disorders in which the defects reside in aspirin. What is lost in these arguments is that endothelial cells
the various intracellular signaling pathways, including defects also produce another potent platelet inhibitor, nitric oxide
in G protein subunits and phospholipase C isoenzymes.25,59,60 (NO), and its production is not affected by aspirin. Although
Scott syndrome is a rare autosomal recessive disorder of aspirin may inhibit a proaggregatory mechanism (thrombox-
calcium-induced membrane phospholipid scrambling (neces- ane production) and an antiaggregatory mechanism (prostacy-
sary for coagulation factor assembly) and thrombin generation clin production), the NO platelet inhibitory mechanism is not
on platelets. Platelets secrete and aggregate normally but do affected. It may be necessary to define a test system to deter-
not transport phosphatidylserine and phosphatidylethanol- mine the optimal dosage of aspirin on an individual basis,
amine from the inner leaflet to the outer leaflet of the plasma because some patients have, or develop, aspirin resistance, and
membrane. This phospholipid flip normally occurs during a dosage that previously was sufficient to inhibit platelet func-
platelet activation and is essential for the binding of vitamin tion effectively may no longer able to produce that effect.
Kdependent clotting factors. In the membrane of resting Finally, unlike the practice with almost all other therapeutic
platelets, phosphatidylserine and phosphatidylethanolamine agents, a single dose of aspirin is usually prescribed in a one
are restricted to the inner leaflet of the plasma membrane, and dose fits all fashion (e.g., 325mg) without regard to the
phosphatidylcholine is expressed on the outer leaflet. This patients weight, age, health status, or other measurable param-
asymmetry is maintained by the enzyme aminophospholipid eters. This practice is based on the assumption that the biologic
translocase.61 When platelets are activated, the asymmetry is effect will be the same in all patients. Evidence is emerging,
lost, and phosphatidylserine and phosphatidylethanolamine however, that there are considerable interindividual differences
flip to the outer leaflet and facilitate the assembly of clotting in the response to a single dose of aspirin.5,25,66-68 One study
factor complexes. The phospholipid flip is mediated by a has shown that patients who do not respond well to aspirin
calcium-dependent enzyme, scramblase.62 In Scott syndrome, have worse outcomes than patients who respond well.69
platelet plug formation (including adhesion, aggregation, and Individuals known to have a defect in their hemostatic
secretion) occurs normally, but clotting factor complexes do mechanism, such as a storage pool deficiency, thrombocytope-
not assemble on the activated platelet surface, and thrombin nia, a vascular disorder, or VWD, may experience a marked
generation is absent or much reduced. Because lack of throm- increase in bleeding time after aspirin ingestion, and such indi-
bin generation leads to inadequate fibrin, the platelet plug is viduals should be advised to avoid the use of aspirin and
not stabilized, and a bleeding diathesis results.63,64 related agents.25
Lastly, Stormorken syndrome is a condition in which plate- The list of drugs affecting the prostaglandin pathway that
lets are always in an activated state and express phosphati- converts arachidonic acid to thromboxane is long and beyond
dylserine on the outer leaflet of the membrane without prior the scope of this chapter. Many of these drugs inhibit cyclooxy-
activation. It has been postulated that patients with this syn- genase, but, unlike with aspirin and closely related compounds,
drome have a defective aminophospholipid translocase.65 the inhibition is reversible. These drugs are said to be competi-
tive inhibitors of cyclooxygenase, and as the blood concentra-
Acquired Defects of Platelet Function tion of the drug decreases, platelet function is recovered. This
Drug-Induced Defects group of drugs includes ibuprofen and related compounds,
Drugs That Inhibit the Prostaglandin Pathway. Unlike such as ketoprofen and fenoprofen, naproxen, and sulfinpyr-
inherited disorders of platelet function, which are rare, acquired azone. In contrast to aspirin, most of these agents have little
disorders of platelet function are commonly encountered. The effect on the bleeding time test. Except for their potential to
most frequent cause of acquired platelet dysfunction is drug irritate the gastric mucosa, these drugs have not been reported
ingestion, with aspirin and other drugs that inhibit the platelet to cause clinically important bleeding.14,17,25,70 Interestingly,
prostaglandin synthetic pathways being the most common ibuprofen appears to have a prothrombotic effect when
culprits. A single 200-mg dose of acetylsalicylic acid (aspirin) ingested within 2 hours of aspirin, because it blocks the acety-
can irreversibly acetylate 90% of the platelet cyclooxygenase lation site for aspirin on COX-1. Patients taking aspirin should
(see Figure 13-19). In platelets the acetylated cyclooxygenase be cautioned to avoid ibuprofen and related drugs near the
(cyclooxygenase-1, or COX-1) is completely inactive. Platelets time of aspirin ingestion.
lack a nucleus and cannot synthesize new enzymes. The inhibi- The association of chronic alcohol consumption with
tory effect is permanent for the circulatory life span of the thrombocytopenia is well known. Chronic, periodic, and even
platelet (7 to 10 days). Endothelial cells synthesize new cyclo- acute alcohol consumption may result in a transient decrease
oxygenase, and endothelial cell cyclooxygenase seems to be less in platelet function, however, and the inhibitory effect seems
sensitive to aspirin than the platelet enzyme, at least at low to be more pronounced when alcohol is consumed in excess.
dosages. This has led to the view that low dosages of aspirin These effects are well known, and most patients who are sched-
may be better than higher dosages, because platelet thrombox- uled to undergo a medical procedure in which there may be
ane production is inhibited whereas endothelial cells recover hemostatic challenge are advised to abstain from alcohol con-
prostacyclin production. Others argue that inhibition of plate- sumption for about 3 days before the procedure. The impaired
let function is the more important effect and that higher platelet function seems to be related at least in part to inhibi-
dosages of aspirin are better for this purpose. For these reasons, tion of thromboxane synthesis. A reduced platelet count and
there are wide-ranging opinions as to the optimal dosage of impaired platelet function may contribute to the increased
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 725
incidence of gastrointestinal hemorrhage associated with agent in this group targets a GP IIb/IIIa recognition site for an
chronic excessive alcohol intake.14,25,71,72 arginineglycineaspartic acid (RGD) sequence found in
fibrinogen and several adhesive proteins. These agents bind to
Drugs That Inhibit Membrane Function. Many drugs the recognition site, prevent the binding of fibrinogen, and
interact with the platelet membrane and cause a clinically sig- consequently prevent platelet aggregation. These compounds
nificant platelet function defect that may lead to hemorrhage. are relatively easily synthesized and contain the RGD sequence
Some of these drugs are useful antiplatelet agents, whereas for recognized by the receptor (tirofiban) or a structure that mimics
many other drugs, their effects on the platelet membrane are the RGD sequence (eptifibatide) and are bound by the RGD
an adverse side effect.50 recognition site on GP IIb/IIIa. When the receptor site is occu-
The thienopyridine derivatives clopidogrel and prasugrel pied by the drug, GP IIb/IIIa is no longer functional as the
and their predecessor ticlopidine are antiplatelet agents used aggregation receptor. The goal of therapy with these drugs is to
to treat patients with arterial occlusive disease for prevention induce a controlled thrombasthenia-like state. At present these
of myocardial infarction, patients with cerebrovascular disease agents are primarily used in patients undergoing percutaneous
for reduction of the risk of thrombotic stroke, and patients who coronary intervention and are administered concurrently with
are intolerant of aspirin. In contrast to the effect of aspirin, the heparin and other antiplatelet agents. Currently, the use of
effects of these agents do not reach a steady state for 3 to 5 these agents is limited by the need to administer them by con-
days, although a steady state can be reached sooner with a stant intravenous infusion. An orally active agent of this type
loading dose. As prophylactic agents, they have been shown to is under development and is in clinical trials.76-78
be as efficacious as aspirin. The mechanism of action seems to Dipyridamole is an inhibitor of platelet phosphodiesterase,
be interference with the binding of ADP to the P2Y12 platelet the enzyme responsible for converting cyclic adenosine mono-
membrane STD receptor.56 The major effect appears to be inhi- phosphate (cAMP) to AMP (see Figure 13-20). Elevation of
bition of stimulus-response coupling between the platelet ADP cytoplasmic cAMP is inhibitory to platelet function, and inhi-
receptor and fibrinogen binding to GP IIb/IIIa. As a conse- bition of phosphodiesterase allows the accumulation of cAMP
quence, platelet activation and aggregation induced by ADP are in the cytoplasm. Dipyridamole alone does not inhibit platelet
markedly inhibited, and responses to other aggregating agents, aggregation in response to the usual platelet agonists, but pro-
such as collagen, are reduced. Clopidogrel has more effect on motes inhibition of agents that stimulate cAMP formation,
the bleeding time than aspirin, although there is little difference such as prostacyclin, stable analogues of prostacyclin, and NO.
in the risk of clinical bleeding.73 Clopidogrel and ticlopidine At one time, dipyridamole, alone or in combination with
can produce major side effects in some patients, including aspirin, was widely used. By the 1990s, interest in dipyridamole
long-lasting neutropenia, aplastic anemia, thrombocytopenia, had waned. There has been a resurgence of interest in dipyri-
gastrointestinal distress, and diarrhea. Clopidogrel has the same damole, however, as a combination agent compounded with
clinical efficacy as ticlopidine, but the incidence and severity of aspirin (Aggrenox).
these side effects are much lower.14,74 Clopidogrel has become Antibiotics are well known for their ability to interfere with
the drug of choice for this class of antiplatelet agents. Clopido- platelet function. Most of the drugs with this effect contain the
grel and aspirin are increasingly being used in combination to -lactam ring and are either a penicillin or a cephalosporin
prevent arterial thrombosis, primarily based on the synergistic (Figure 44-2). These drugs can prolong the bleeding time, but
action of these two drugs, which inhibit platelet function by this effect is seen only in patients receiving large parenteral
different mechanisms. Similar to aspirin effects, the effects of
the thienopyridines are not readily reversible; platelet function
after cessation of drug intake is about 50% of normal at 3 days,
and complete recovery of function occurs at 7 to 10 days.75
A newer group of antiplatelet agents targets the platelet
membrane GP IIb/IIIa (IIb/3) receptor, interfering with the
ability of this receptor to bind fibrinogen and inhibiting plate-
let aggregation in response to all of the usual platelet aggregat-
ing agents. Results of platelet function studies on platelets
from patients receiving therapeutic dosages of these drugs
essentially mimic those of a mild form of Glanzmann throm-
basthenia. At present, three of these agents are approved for
use in the United States. Two different types of agents are
included in this group. The first such agent approved for clini-
cal use in the United States was the Fab fragment of the mouse/
human chimeric monoclonal antibody 7E3 (c7E3 Fab; abcix-
imab [ReoPro]), which binds to GP IIb/IIIa, prevents the
binding of fibrinogen, and prevents platelet aggregation. Figure 44-2 Chemical structure of major classes of -lactam antibiotics.
Numerous studies have shown the efficacy of this drug as R, Nonspecified side chain. (From Mahon CR, Lehman DE, Manuselis G:
an antiplatelet and antithrombotic agent. The second type of Textbook of diagnostic microbiology, ed 4, St Louis, 2011, Saunders.)
726 PART VIII Hemostasis and Thrombosis

doses and is a problem only for hospitalized patients. One myelogenous leukemia, essential thrombocythemia, and
postulated mechanism for the antiplatelet effect of these drugs myelofibrosis with myeloid metaplasia (see Chapter 34). Plate-
is that they associate with the membrane via a lipophilic reac- let dysfunction is a common finding in patients with these
tion and block receptor-agonist interactions or stimulus- disorders. Hemorrhagic complications occur in about one
response coupling. They also may inhibit calcium influx in third, thrombosis occurs in another third, and although it is
response to thrombin stimulation, reducing the ability of uncommon, both develop in some patients. These complica-
thrombin to activate platelets. Although these drugs may tions are serious causes of morbidity and mortality. Although
prolong the bleeding time and in vitro aggregation responses the occurrence of hemorrhage or thrombosis in MPN patients
to certain agonists, their association with a hemostatic defect is largely unpredictable, certain patterns have emerged. Hemor-
severe enough to cause clinical hemorrhage is uncertain and is rhage and thrombosis are less common in chronic myeloge-
not predicted by the bleeding time test results.5,17,51,74 nous leukemia than in the other MPNs. Bleeding seems to be
Nitrofurantoin is an antibiotic that is not related to the more common in myelofibrosis with myeloid metaplasia, but
-lactam drugs but may prolong the bleeding time and inhibit thrombosis is more common in the other MPNs. Abnormal
platelet aggregation when high concentrations are present in platelet function has been postulated as a contributing cause.
the blood. This drug is not known to cause clinical bleeding, This hypothesis is supported by the observation that bleeding
however.74 is usually mucocutaneous in nature, and thrombosis may be
The dextrans, another class of commonly used drugs, can arterial or venous. In patients with these disorders, thrombosis
prolong the bleeding time, inhibit platelet aggregation, and may occur in unusual sites, including the mesenteric, hepatic,
impair platelet procoagulant activity when given as an intrave- and portal circulations. Patients with essential thrombocythe-
nous infusion. These drugs have no effect on platelet function, mia may develop digital artery thrombosis and ischemia of the
however, when added directly to platelet-rich plasma. Dextrans fingers and toes, occlusions of the microvasculature of the
are partially hydrolyzed, branched-chain polysaccharides of heart, and cerebrovascular occlusions that result in neurologic
glucose. The two most commonly used are dextran 70 (molecu- symptoms.74
lar mass of 70,000 to 75,000 D) and dextran 40 (molecular In MPNs, a variety of platelet function defects have been
mass of 40,000 D), also known as low-molecular-weight dextran. described, but their clinical importance is uncertain. Platelets
Both drugs are effective plasma expanders and are commonly have been reported to have abnormal shapes, decreased proco-
used for this purpose. Because of their effects on platelets, they agulant activity, and a decreased number of secretory granules.
have been extensively used as antithrombotic agents. There In essential thrombocythemia, platelet survival may be short-
does not seem to be any increased risk of hemorrhage associ- ened. The bleeding time is prolonged in only a few patients,
ated with the use of these agents, but their efficacy in prevent- and hemorrhage can occur in patients with a normal bleeding
ing postoperative pulmonary embolism is equal to that of time. The risk of thrombosis or hemorrhage correlates poorly
low-dose subcutaneous heparin.14,44,71,74 with the elevation of the platelet count.
Hydroxyethyl starch, or hetastarch, is a synthetic glucose The most common abnormalities are decreased aggregation
polymer with a mean molecular mass of 450,000 D that also and secretion in response to epinephrine, ADP, and collagen.79
is used as a plasma expander. It has effects similar to those of Possible causes of the platelet dysfunction include loss of plate-
the dextrans. The mechanism of action of these drugs has not let surface membrane -adrenergic (epinephrine) receptors,
been clearly elucidated but is presumed to involve interaction impaired release of arachidonic acid from membrane phos-
with the platelet membrane.14,44,71 pholipids in response to stimulation by agonists, impaired
oxidation of arachidonic acid by the cyclooxygenase and lipox-
Miscellaneous Drugs That Inhibit Platelet Function. ygenase pathways, a decrease in the contents of -granules and
Several other agents of diverse chemical structure and function -granules, and loss of a variety of platelet membrane receptors
are known to inhibit platelet function. The mechanisms by for adhesion and activation. There seems to be no correlation
which they induce platelet dysfunction are largely unknown. between a given MPN and the type of platelet dysfunction
Nitroglycerin, nitroprusside, propranolol, and isosorbide dini- observed, with the exception that most patients with essential
trate are drugs used to regulate cardiovascular function that thrombocythemia lack an in vitro platelet aggregation response
seem to be able to cause a decrease in platelet secretion and to epinephrine. This observation may be helpful in the differ-
aggregation. Patients taking phenothiazine or tricyclic anti ential diagnosis.14,17,74,80
depressants may have decreased secretion and aggregation
responses, but these effects are not associated with an increased Multiple Myeloma and Waldenstrm Macroglobu
risk for hemorrhage. Local and general anesthetics may impair linemia. Platelet dysfunction is observed in approximately
in vitro aggregation responses. The same is true of antihista- one third of patients with IgA myeloma or Waldenstrm macro
mines. Finally, some radiographic contrast agents are known globulinemia, a much smaller percentage of patients with IgG
to inhibit platelet function.74 multiple myeloma, and only occasionally in patients with
monoclonal gammopathy of undetermined significance (see
Disorders That Affect Platelet Function Chapter 37). Platelet dysfunction results from coating of the
Myeloproliferative Neoplasms. Chronic myeloprolifer platelet membranes by paraprotein and does not depend on
ative neoplasms (MPNs) include polycythemia vera, chronic the type of paraprotein present. In addition to interacting with
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 727
platelets, the paraprotein may interfere with fibrin polymeriza- of alcohol on bone marrow megakaryocytes. The severe bleed-
tion and the function of other coagulation proteins. Almost all ing diathesis associated with end-stage liver disease has many
patients with malignant paraprotein disorders have clinically causes, such as markedly decreased or negligible coagulation
significant bleeding, but thrombocytopenia is still the most factor production, excessive fibrinolysis, dysfibrinogenemia,
likely cause of bleeding in these patients. Other causes of thrombocytopenia, and (occasionally) disseminated intra
bleeding include hyperviscosity syndrome, complications of vascular coagulation. Upper gastrointestinal tract bleeding is a
amyloidosis (e.g., acquired factor X deficiency), and, in rare relatively common feature of cirrhosis, particularly alcoholic
instances, presence of a circulating heparin-like anticoagulant cirrhosis, and recombinant factor VIIa has been shown to be
or fibrinolysis.16,71,74 effective treatment in some patients.81

Cardiopulmonary Bypass Surgery. Cardiopulmonary Uremia. Uremia is commonly accompanied by bleeding


bypass induces thrombocytopenia and a severe platelet func- caused by platelet dysfunction. In uremia, guanidinosuccinic
tion defect that assumes major importance in surgical bleeding acid (GSA) is present in the circulation in higher than normal
after bypass. The function defect most likely results from plate- levels as a result of inhibition of the urea cycle. GSA is dialyz-
let activation and fragmentation in the extracorporeal circuit. able, and dialysis (peritoneal dialysis or hemodialysis) is
Causes of platelet activation include adherence and aggregation usually effective in correcting the prolonged bleeding time and
of platelets to fibrinogen (adsorbed onto the surfaces of the the abnormal platelet function characteristic of uremia. NO
bypass circuit material), mechanical trauma and shear stresses, diffuses into platelets, activates soluble guanylate cyclase,
blood conservation devices, bypass pump-priming solutions, and inhibits platelet adhesion, activation, and aggregation.82
hypothermia, complement activation, and exposure of platelets Because GSA is an NO donor, NO is present in the circulation
to the blood-air interface in bubble oxygenators. Some degree at higher than normal levels in uremia. Abnormal platelet func-
of platelet degranulation typically is found after bypass surgery, tion is far more common than clinically significant bleeding in
which indicates that platelet activation and secretion has uremic patients.2,50,52
occurred during the operation. Platelet membrane fragments Platelet aggregation pattern abnormalities are not uniform,
or microparticles are found consistently in the blood of and any combination of defects may be seen. There is evidence
bypass patients, providing additional evidence of the severe of a deficient release reaction, such as lack of primary ADP-
mechanical stress encountered by platelets during bypass pro- induced aggregation, and subnormal platelet procoagulant
cedures. The severity of the platelet function defect closely activity. The bleeding time is characteristically prolonged in
correlates with the length of time on bypass. After an uncom- uremia and seems to correlate with the severity of renal failure
plicated bypass procedure, normal platelet function returns in in these patients. There does not seem to be any significant
about 1 hour, although the platelet count does not return to correlation, however, between the bleeding time and the risk
normal for several days. Thrombocytopenia is caused by hemo- of clinically significant bleeding. Anemia is an independent
dilution, accumulation of platelets on the surfaces of the bypass cause of prolonged bleeding time, and the severity of anemia
materials, sequestration or removal of damaged platelets by the in uremic patients correlates with the severity of renal failure.
liver and reticuloendothelial system, and consumption associ- Many uremic patients are treated with recombinant erythro
ated with normal hemostatic processes after surgery.71,74 poietin to increase their hematocrit. Maintenance of the hema-
tocrit at greater than 30% also may help to normalize the
Liver Disease. Moderate to severe liver disease is reported bleeding times.2,71,73
to be associated with a variety of hemostatic abnormalities, Bleeding is uncommon in uremic patients and is seen more
including reduction in clotting proteins, reduction of proteins often with concurrent use of drugs that interfere with platelet
in the natural anticoagulant pathways, dysfibrinogenemia, and function or in association with heparin use in hemodialysis.
excessive fibrinolysis. Mild to moderate thrombocytopenia is Platelet concentrates often are used to treat severe hemorrhagic
seen in approximately one third of patients with chronic liver episodes in patients with uremia but usually do not correct
disease in association with hypersplenism or as a result of the bleeding. Other therapies that are sometimes effective
alcohol toxicity.4,74 include cryoprecipitate, desmopressin acetate, and conjugated
Abnormal platelet function test results seen in patients with estrogen.2,71,73
chronic liver disease include reduced platelet adhesion; abnor-
mal platelet aggregation in response to ADP, epinephrine, and Hereditary Afibrinogenemia. Hereditary afibrinogen-
thrombin; abnormal platelet factor 3 availability; and reduced emia has been documented in more than 150 families.
procoagulant activity. An acquired storage pool deficiency also Although it is not truly a platelet function disorder, platelets
has been suggested. The prolonged bleeding time in these do not exhibit normal function in the absence or near-absence
patients may respond to infusion of desmopressin acetate. It is of fibrinogen. In most patients, the bleeding time is prolonged,
unclear, however, whether desmopressin acetate provides a and because fibrinogen is essential for normal platelet aggrega-
benefit in preventing bleeding in these patients or is simply tion, platelet aggregation test results are abnormal. Abnormal
correcting an abnormal laboratory test result. results on platelet retention and adhesion studies involving the
In chronic alcoholic cirrhosis, the thrombocytopenia and use of glass beads also have been documented. In addition,
platelet abnormalities may result from the direct toxic effects results of all clot-based tests (including partial thromboplastin
728 PART VIII Hemostasis and Thrombosis

time, prothrombin time, reptilase time, thrombin time, and


whole-blood clotting time) are abnormal. Addition of fibrino- BOX 44-2 Vascular Disorders
gen to samples or infusion of fibrinogen into the patient results Hereditary vascular disorders
in correction of the abnormal test results.2,83 Hereditary hemorrhagic telangiectasia (Rendu-Osler-Weber syndrome)
A high incidence of hemorrhagic manifestations is found Hemangioma-thrombocytopenia syndrome (Kasabach-Merritt syn-
in patients with afibrinogenemia (or severe hypofibrinogen- drome)
emia). Bleeding is the cause of death in about one third of Ehlers-Danlos syndrome and other genetic disorders
such patients. Cryoprecipitate or fibrinogen concentrates can Acquired vascular disorders
be used to treat bleeding episodes. Some patients develop Allergic purpura (Henoch-Schnlein purpura)
antibodies to fibrinogen, and this treatment then becomes Senile purpura
ineffective.83 Paraproteinemia and amyloidosis
Drug-induced vascular purpuras
Hyperaggregable Platelets Vitamin C deficiency (scurvy)
Patients with a variety of disorders associated with thrombosis Purpuras of unknown origin
or increased risk for thrombosis, including hyperlipidemia, Purpura simplex (easy bruisability)
diabetes mellitus, peripheral arterial occlusive disease, acute Psychogenic purpura
arterial occlusion, myocardial infarction, and stroke, have From Thompson AR, Harker LA: Manual of hemostasis and thrombosis, ed 3,
been reported to have increased platelet reactivity. Platelets Philadelphia, 1983, FA Davis.
from these patients tend to aggregate at lower concentrations
of aggregating agents than do platelets from individuals
without these conditions. Spontaneous aggregation (aggre
gation in response to stirring only) is also an indicator of Hereditary Vascular Disorders
abnormally increased platelet reactivity and often accompanies Hereditary Hemorrhagic Telangiectasia
increased sensitivity to platelet agonists. The presence of (Rendu-Osler-Weber Syndrome)
spontaneous aggregation by itself is considered to be consistent The mode of inheritance of hereditary hemorrhagic telangi-
with the presence of a hyperaggregable state. Because partici ectasia is autosomal dominant. The vascular defect of this
pation of platelets is necessary for the development of disorder is characterized by thin-walled blood vessels with a
arterial thrombosis, the presence of hyperaggregable platelets discontinuous endothelium, inadequate smooth muscle, and
is often an indication that an antiplatelet agent should be used inadequate or missing elastin in the surrounding stroma.
as part of a therapeutic or prophylactic regimen for arterial Telangiectasias (dilated superficial blood vessels that create
thrombosis.71,84,85 small, focal red lesions) occur throughout the body but are
Acquired platelet function defects are seen occasionally in most obvious on the face, lips, tongue, conjunctiva, nasal
patients with autoimmune disorders, including systemic lupus mucosa, fingers, toes, and trunk and under the tongue. The
erythematosus, rheumatoid arthritis, scleroderma, and the lesions blanch when pressure is applied. The disorder usually
immune thrombocytopenias, such as immune thrombocyto- becomes manifest by puberty and progresses throughout life.
penic purpura.74 Telangiectasias are fragile and prone to rupture. Epistaxis is
Purified fibrin degradation products can induce platelet dys- an almost universal finding, and symptoms almost always
function in vitro. The pathophysiologic relevance of this obser- worsen with age. The age at which nosebleeds begin is
vation is uncertain, because the concentrations of fibrin a good gauge of the severity of the disorder. Although the
degradation products required are unlikely to be reached in oral cavity, gastrointestinal tract, and urogenital tract are
vivo. Patients with disseminated intravascular coagulation may common sites of bleeding, bleeding can occur in virtually
have reduced platelet function, however, as a result of in vivo every organ.87
stimulation by thrombin and other agonists, resulting in in The laboratory findings relate to the severity of the hemor-
vivo release of granule contents. This has been called acquired rhagic tendencies. The bleeding time is usually normal, and
storage pool disease; the term exhausted platelets may be more the tourniquet test result may be normal or show increased
appropriate.86 capillary fragility. The diagnosis of hereditary hemorrhagic tel-
angiectasia is based on the characteristic skin or mucous mem-
brane lesions, a history of repeated hemorrhage, and a family
VASCULAR DISORDERS
history of a similar disorder. Patients with hereditary hemor-
The pathophysiology of disorders of vessels and their support- rhagic telangiectasia do well despite the lack of specific therapy
ing tissues is obscure. Laboratory studies of platelets and blood and the seriousness of their hemorrhagic manifestations.2,6,87
coagulation usually yield normal results. The diagnosis is often There are several other disorders and conditions in which tel-
based on medical history and is made by ruling out other angiectasias are present, including cherry-red hemangiomas
sources of bleeding disorders. The usual clinical sign is the (common in older men and women), ataxia-telangiectasia
tendency to bruise easily or to bleed spontaneously, especially (Louis-Bar syndrome), and chronic actinic telangiectasia; they
from mucosal surfaces. Vascular disorders are summarized in also are seen in association with chronic liver disease and
Box 44-2. pregnancy.87
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 729
Hemangioma-Thrombocytopenia Syndrome affected. The onset of the disease is sudden, often following an
(Kasabach-Merritt Syndrome) upper respiratory tract infection. The infectious organism may
Kasabach and Merritt originally described the association of a damage the endothelial lining of blood vessels, which results
giant cavernous hemangioma (vascular tumor), thrombocyto- in vasculitis. Attempts have been made to implicate a specific
penia, and a bleeding diathesis. The hemangiomas are visceral infectious agent, particularly -hemolytic streptococcus.2,6
or subcutaneous, but rarely both. External hemangiomas may Malaise, headache, fever, and rash may be the presenting
become engorged with blood and resemble hematomas. Other symptoms. The delay in the appearance of the skin rash often
well-recognized features of Kasabach-Merritt syndrome include poses a difficult problem in differential diagnosis. The skin
acute or chronic disseminated intravascular coagulation and lesions are urticarial and gradually become pinkish, then red,
microangiopathic hemolytic anemia. A hereditary basis for this and finally hemorrhagic. The appearance of the lesions may be
syndrome has not been established, but the condition is very rapid and accompanied by itching. The lesions have been
present at birth. Several treatment modalities are available for described as palpable purpura, in contrast to the perfectly flat
the tumors and the coagulopathy and range from cortico lesions of thrombocytopenia and most other forms of vascular
steroid therapy to surgery.6,88 purpura. These lesions are most commonly found on the feet,
elbows, knees, buttocks, and chest. Ultimately, a brownish red
Ehlers-Danlos Syndrome and eruption is seen. Petechiae also may be present.2,6
Other Genetic Disorders As the disease progresses, abdominal pain, polyarthralgia,
Ehlers-Danlos syndrome may be transmitted as an autosomal headaches, and renal disease may develop. Renal lesions are
dominant, recessive, or X-linked trait. It is manifested by hyper- present in 60% of patients during the second to third week
extensible skin, hypermobile joints, joint laxity, fragile tissues, of the disorder. Proteinuria and hematuria are commonly
and a bleeding tendency, primarily subcutaneous hematoma present.2,4,6
formation. Eleven distinct varieties of the disorder are recog- The platelet count is normal. Tests of hemostasis, including
nized. The severity of bleeding ranges from easy bruisability to the bleeding time, tourniquet test, and tests of blood coagula-
arterial rupture. The disorder generally can be ascribed to tion, usually yield normal results in patients with allergic
defects in collagen production, structure, or cross-linking, with purpura. Anemia generally is not present unless the hemor-
resulting inadequacy of the connective tissues. Platelet abnor- rhagic manifestations have been severe. The white blood cell
malities have been reported in some patients. Common abnor- count and the erythrocyte sedimentation rate are usually ele-
mal laboratory findings include a positive tourniquet test result vated. The disease must be distinguished from other forms of
and prolonged bleeding time.6 nonthrombocytopenic purpura. Numerous infectious diseases
Other inherited vascular disorders include pseudoxanthoma that may be associated with purpura also must be considered
elasticum and homocystinuria (autosomal recessive disorders), in the differential diagnosis. Drugs or chemicals sometimes
and Marfan syndrome and osteogenesis imperfecta (autosomal may be implicated.2,6
dominant disorders). In addition to vascular defects, Marfan In the pediatric age group, the average duration of the initial
syndrome is characterized by skeletal and ocular defects.6 episode is about 4 weeks. Relapses are frequent, usually after a
period of apparent well-being. Except for patients in whom
Acquired Vascular Disorders chronic renal disease develops, the prognosis is usually good.
Allergic Purpura (Henoch-Schnlein Purpura) Occasionally, death from renal failure has occurred. Manage-
The term allergic purpura or anaphylactoid purpura generally is ment is directed primarily at symptomatic relief, because there
applied to a group of nonthrombocytopenic purpuras charac- currently is no effective treatment. Corticosteroids sometimes
terized by apparently allergic manifestations, including skin have been helpful in alleviating symptoms. Most patients
rash and edema. Allergic purpura has been associated with recover without treatment.2,6
certain foods and drugs, cold, insect bites, and vaccinations.
The term Henoch-Schnlein purpura is applied when the con Paraproteinemia and Amyloidosis
dition is accompanied by transient arthralgia, nephritis, abdo Platelet function can be inhibited by myeloma proteins. Abnor-
minal pain, and purpuric skin lesions, which are frequently malities in platelet aggregation, secretion, clot retraction, and
confused with the hemorrhagic rash of immune thrombocyto- procoagulant activity correlate with the concentration of the
penic purpura.2,6,44 plasma paraprotein and are likely due to coating of the platelet
General evidence implicates autoimmune vascular injury, membrane with the paraprotein. Under these conditions, plate-
but the pathophysiology of the disorder is unclear. Preliminary let adhesion and activation receptor functions are inhibited,
evidence indicates that the vasculitis is mediated by immune and the paraprotein coating also inhibits assembly of clotting
complexes containing IgA antibodies. It has been suggested factors on the platelet surface. High concentrations of parapro-
that allergic purpura may represent autoimmunity to compo- tein can cause severe hemorrhagic manifestations as a result of
nents of vessel walls.2,6 a combination of hyperviscosity and platelet dysfunction.
Henoch-Schnlein purpura is primarily a disease of chil- About one third of patients with IgA myeloma and Walden-
dren, occurring most commonly in children 3 to 7 years of age. strm macroglobulinemia and approximately 5% of patients
It is relatively uncommon among individuals younger than with IgG myeloma (usually IgG3) exhibit platelet function
age 2 and older than age 20. Twice as many boys as girls are abnormalities. Finally, the paraprotein may contribute further
730 PART VIII Hemostasis and Thrombosis

to bleeding by inhibiting fibrin polymerization. In these in the skin (age spots). The lesions are limited mostly to the
patients, there is poor correlation between abnormal results on extensor surfaces of the forearms and backs of the hands and
laboratory tests (e.g., prothrombin time, activated partial occasionally occur on the face and neck. With the exception
thromboplastin time, thrombin time, bleeding time) and evi- of increased capillary fragility, results of laboratory tests are
dence of clinical bleeding. Treatment for the bleeding complica- normal, and no other bleeding manifestations are present.2,6
tions of these disorders is primarily reduction in the level of
the paraprotein. This can be accomplished quickly, albeit tran- Drug-Induced Vascular Purpuras
siently, by plasmapheresis. Longer-term treatment is usually Purpura associated with drug-induced vasculitis occurs in the
chemotherapy for the underlying plasma cell malignancy.74,89 presence of functionally adequate platelets. A variety of drugs
Amyloid is a fibrous protein consisting of rigid, linear, non- are known to cause vascular purpura, including aspirin, warfa-
branching, aggregated fibrils approximately 7.5 to 10nm wide rin, barbiturates, diuretics, digoxin, methyldopa, and several
and of indefinite length. Amyloid is deposited extracellularly antibiotics. Sulfonamides and iodides have been implicated
and may lead to damage of normal tissues. Various proteins most frequently. The lesions vary from a few petechiae to
can serve as subunits of the fibril, including monoclonal light massive, generalized petechial eruptions. Mechanisms include
chains ( more frequently than ). Amyloidosis, the deposition development of antibodies to vessel wall components, develop-
of abnormal quantities of amyloid in tissues, may be primary ment of immune complexes, and changes in vessel wall perme-
or secondary and localized or systemic. A discussion of the ability. As soon as the disorder is recognized, the offending drug
clinical spectrum of amyloidosis is beyond the scope of this should be discontinued. No other treatment is necessary.6
chapter. Purpura, hemorrhage, and thrombosis may be a part
of the clinical presentation of patients with amyloidosis, Miscellaneous Causes of Vascular Purpura
however. Thrombosis and hemorrhage have been ascribed to Insufficient dietary intake of vitamin C (ascorbic acid) results
amyloid deposition in the vascular wall and surrounding in scurvy and decreased synthesis of collagen, with weakening
tissues. Platelet function has been shown to be abnormal in a of capillary walls and the appearance of purpuric lesions.6 A
few cases, and in rare cases patients may have thrombocyto diagnosis of purpura simplex (simple vascular purpura) or
penia. Current treatments for amyloidosis are not effective.90 vascular fragility is made when a cause for purpura cannot be
found. The ecchymoses are superficial, bleeding is usually
Senile Purpura mild, and laboratory test results are most often normal.6 Cuta-
Senile purpura occurs more commonly in elderly men than in neous bleeding and bruising through intact skin has been
women and is due to a lack of collagen support for small blood observed in patients in whom no vascular or platelet dysfunc-
vessels and loss of subcutaneous fat and elastic fibers. The tion can be detected. Most such cases involve women with
incidence increases with advancing age. The dark blotches are emotional problems, and the bruising is often accompanied by
flattened, are about 1 to 10mm in diameter, do not blanch nausea, vomiting, or fever. Evidence for a psychosomatic origin
with pressure, and resolve slowly, often leaving a brown stain is equivocal. Laboratory test results are invariably normal.6

SUMMARY
Inherited qualitative platelet disorders can cause bleeding disor- Drugs are the most common cause of acquired platelet dysfunc-
ders ranging from mild to severe. tion, and aspirin is the most frequent culprit. Several new classes
BSS is caused by the lack of expression of GP Ib/IX/V complexes on of antiplatelet agents with different effects than aspirin are now
the platelet surface. This receptor complex is responsible for plate- available and gaining in popularity.
let adhesion, and its absence results in a severe bleeding disorder. A variety of pathologic conditions can result in platelet dysfunction
Glanzmann thrombasthenia is caused by the lack of expression and range from hematologic malignancies to kidney disease and
of GP IIb/IIIa complexes on the platelet surface. This complex is liver disease.
known as the platelet aggregation receptor, and its absence is Vascular disorders that result in bleeding are uncommon. There
associated with a severe bleeding disorder. are a few well-recognized inherited disorders, however, such as
Storage pool disorders result from the absence of intraplatelet Ehlers-Danlos syndrome and hereditary hemorrhagic telangiecta-
-granules, dense granules, or both. Platelet dysfunction associ- sia, that can result in substantial blood loss.
ated with these disorders is generally mild; bleeding symptoms Vascular disorders can be acquired, and these are much more
also are usually mild. common than inherited disorders. Causes range from the effects
Aspirin-like effects result from defects in elements of the arachi- of aging to drug effects to allergic reaction.
donic acid metabolic pathway. Platelet dysfunction mimics that
seen after aspirin ingestion. Now that you have completed this chapter, go back and
Deficiencies of several of the receptors for platelet-activating read again the case study at the beginning and respond
substances have been documented, and bleeding symptoms of to the questions presented.
varying severity are associated with these deficiencies.
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 731

R E V I E W Q UESTIONS
1. The clinical presentation of platelet-related bleeding may 7. The impaired platelet function in MPNs results from:
include all of the following except: a. Abnormally shaped platelets
a. Bruising b. Extended platelet life span
b. Nosebleeds c. Increased procoagulant activity
c. Gastrointestinal bleeding d. Decreased numbers of - and -granules
d. Bleeding into the joints (hemarthroses)
8. Which is a congenital qualitative platelet disorder?
2. A defect in GP IIb/IIIa causes: a. Senile purpura
a. Glanzmann thrombasthenia b. Ehlers-Danlos syndrome
b. BSS c. Henoch-Schnlein purpura
c. Gray platelet syndrome d. Waldenstrm macroglobulinemia
d. Storage pool disease
9. In uremia, platelet function is impaired by higher than
3. Aspirin ingestion blocks the synthesis of: normal levels of:
a. Thromboxane A2 a. Urea
b. Ionized calcium b. Uric acid
c. Collagen c. Creatinine
d. ADP d. NO

4. Patients with BSS have which of the following laboratory 10. The platelet defect associated with increased paraproteins
test findings? is:
a. Abnormal platelet response to arachidonic acid a. Impaired membrane activation owing to protein
b. Abnormal platelet response to ristocetin coating
c. Abnormal platelet response to collagen b. Hypercoagulability owing to antibody binding and
d. Thrombocytosis membrane activation
c. Impaired aggregation because the hyperviscous
5. Which of the following is the most common of the heredi- plasma prevents platelet-endothelium interaction
tary platelet function defects? d. Hypercoagulability because the increased proteins
a. Glanzmann thrombasthenia bring platelets closer together, which lead to
b. BSS inappropriate aggregation
c. Storage pool defects
d. Multiple myeloma

6. A mechanism of antiplatelet drugs targeting GP IIb/IIIa


function is:
a. Interference with platelet adhesion to the
subendothelium by blocking of the collagen binding
site
b. Inhibition of transcription of the GP IIb/IIIa gene
c. Direct binding to GP IIb/IIIa
d. Interference with platelet secretion

REFERENCES
1. Triplett DA: How to evaluate platelet function. Lab Med Pract 5. Thompson AR, Harker LA: Manual of hemostasis and throm
Phys July-Aug:37-43, 1978. bosis, ed 3, Philadelphia, 1983, FA Davis.
2. Thorup OA: Leavell and Thorups fundamentals of clinical hema- 6. Powers LW: Diagnostic hematology: clinical and technical prin-
tology, ed 5, Philadelphia, 1987, Saunders. ciples, St Louis, 1989, Mosby.
3. Geddis AE, Kaushansky K: Inherited thrombocytopenias: 7. Nurden P, Nurden A: Giant platelets, megakaryocytes and the
toward a better molecular understanding of disorders of expression of glycoprotein Ib-IX complexes. C R Acad Sci III
platelet production. Curr Opin Pediatr 16:15-24, 2004. 319:717-726, 1996.
4. Coller BS, Mitchell WB, French DL: Hereditary qualitative 8. Clementson KJ: Platelet GP Ib-V-IX complex. Thromb Haemost
platelet disorders. In Lichtman MA, Beutler E, Kipps TJ, et al, 78:266-270, 1997.
editors: Williams hematology, ed 7, New York, 2006, McGraw- 9. Nurden AT: Inherited abnormalities of platelets. Thromb
Hill, pp 1795-1832. Haemost 82:468-480, 1999.
732 PART VIII Hemostasis and Thrombosis

10. Lopez JA, Andrews RK, Afshar-Kharghan V, et al: Bernard- 30. Caen JP: Glanzmanns thrombasthenia. Clin Hematol 1:383-
Soulier syndrome. Blood 91:4397-4418, 1998. 392, 1972.
11. Takahashi H, Murata M, Moriki T, et al: Substitution of Val 31. George JN, Caen JP, Nurden AT: Glanzmanns thrombasthe-
for Met at residue 239 of platelet glycoprotein Ib alpha in nia: the spectrum of clinical disease. Blood 75:1383-1395,
Japanese patients with platelet-type von Willebrand disease. 1990.
Blood 85:727-733, 1995. 32. Jennings LK, Wang WC, Jackson CW, et al: Hemostasis in
12. Matsubara Y, Murata M, Sugita K, et al: Identification of a Glanzmanns thrombasthenia (GT): GT platelets interfere
novel point mutation in platelet glycoprotein Ib, Gly to Ser with the aggregation of normal platelets. Am J Pediatr Hematol
at residue 233, in a Japanese family with platelet-type von Oncol 13:84-90, 1991.
Willebrand disease. J Thromb Haemost 1:2198-2205, 2003. 33. Poon MC, dOiron R, Von Depka M, et al: International Data
13. Othman M, Elbatarny HS, Notley C, et al: Identification and Collection on Recombinant Factor VIIa and Congenital
functional characterization of a novel 27bp deletion in Platelet Disorders Study Group: Prophylactic and therapeutic
the macroglycopeptide-coding region of the GPIb gene recombinant factor VIIa administration to patients with
resulting in platelet-type von Willebrand disease. Blood 104 Glanzmanns thrombasthenia: results of an international
(abstract 1023). 2004. survey. J Thromb Haemost 2:1096-1103, 2004.
14. George JN, Shattil SJ: The clinical importance of acquired 34. Lisman T, Adelmaier J, Heijnen HFG, et al: Recombinant
abnormalities of platelet function. N Engl J Med 324:27-39, factor VIIa restores aggregation of IIb/3-deficient platelets
1991. via tissue factor-independent fibrin generation. Blood 103:
15. Caen JP: Glanzmanns thrombasthenia. Baillieres Clin Haema- 1720-1727, 2004.
tol 2:609-623, 1989. 35. Pati H, Saraya AK: Platelet storage pool disease. Indian J Med
16. Triplett DA, editor: Platelet function: laboratory evaluation and Res 84:617-620, 1986.
clinical application, Chicago, 1978, American Society of Clini- 36. Rao AK: Hereditary disorders of platelet secretion and signal
cal Pathologists. transduction. In Colman RW, Marder VJ, Clowes AW, et al,
17. Rapaport SI: Introduction to hematology, ed 2, Philadelphia, editors: Hemostasis and thrombosis: basic principles and clinical
1987, JB Lippincott. practice, ed 5, Philadelphia, 2006, Lippincott Williams &
18. Meyer M, Kirchmaier CM, Schirmer A, et al: Acquired disor- Wilkins, pp 961-974.
der of platelet function associated with autoantibodies 37. Weiss HJ, Lages B, Vicic W, et al: Heterogeneous abnormali-
against membrane glycoprotein IIb-IIIa complex: 1. Glyco- ties of platelet dense granule ultrastructure in 20 patients
protein analysis. Thromb Haemost 65:491-496, 1991. with congenital storage pool deficiency. Br J Haematol 83:282-
19. Tarantino MD, Corrigan JJ, Jr, Glasser L, et al: A variant 295, 1993.
form of thrombasthenia. Am J Dis Child 145:1053-1057, 38. Bennett JS: Hereditary disorders of platelet function. In
1991. Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology:
20. Friedlander M, Brooks PC, Shaffer RW, et al: Definition of basic principles and practice, ed 4, Philadelphia, 2005, Churchill
two angiogenic pathways by distinct av integrins. Science Livingstone, pp 2327-2345.
270:1500-1502, 1995. 39. Abrams CS, Brass LF: Platelet signal transduction. In Colman
21. Nurden AT, George JN: Inherited disorders of the platelet RW, Marder VJ, Clowes AW, et al, editors: Hemostasis and
membrane: Glanzmann thrombasthenia, Bernard-Soulier thrombosis: basic principles and clinical practice, ed 5, Philadel-
syndrome and other disorders. In Colman RW, Marder VJ, phia, 2006, Lippincott Williams & Wilkins, pp 617-630.
Clowes AW, et al, editors: Hemostasis and thrombosis: basic 40. Jedlitschky G, Tirwschmann K, Lubenow LE, et al: The nucle-
principles and clinical practice, ed 5, Philadelphia, 2006, otide transporter MRP4 (ABCC4) is highly expressed in
Lippincott Williams & Wilkins, pp 987-1010. human platelets and present in dense granules, indicating a
22. Nurden AT: Qualitative disorders of platelets and megakaryo- role in mediator storage. Blood 104:3603-3610, 2004.
cytes. J Thromb Haemost 3:1773-1782, 2005. 41. Spritz RA: Molecular genetics of the Hermansky-Pudlak and
23. Coller BS, Cheresh DA, Asch E, et al: Platelet vitronectin Chdiak-Higashi syndromes. Platelets 9:21-29, 1998.
receptor expression differentiates Iraqi-Jewish from Arab 42. DellAngellica EC, Aguilar RC, Wolins N, et al: Molecular
patients with Glanzmann thrombasthenia in Israel. Blood characterization of the protein encoded by the Hermansky-
77:75-83, 1991. Pudlak syndrome type 1 gene. J Biol Chem 275:1300-1306,
24. French DL: The molecular genetics of Glanzmanns throm- 2000.
basthenia. Platelets 9:5-20, 1998. 43. White JG: Inherited abnormalities of the platelet membrane
25. Corriveau DM, Fritsma GA: Hemostasis and thrombosis in the and secretory granules. Hum Pathol 18:123-139, 1987.
clinical laboratory, Philadelphia, 1988, JB Lippincott. 44. Quick AJ: Hemorrhagic diseases and thrombosis, ed 2, Philadel-
26. Rosa JP, Artanuthurry V, Grelac F, et al: Reassessment of phia, 1966, Lea & Febiger.
protein tyrosine phosphorylation in thrombasthenic plate- 45. Cleary AM, Insel RA, Lewis DB: Disorders of lymphocyte
lets: evidence that phosphorylation of cortactin and a 64kD function. In Hoffman R, Benz EJ, Jr, Shattil SJ, et al, editors:
protein is dependent on thrombin activation and integrin Hematology: basic principles and practice, ed 4, Philadelphia,
IIb3. Thromb Haemost 89:4385-4392, 1997. 2005, Churchill Livingstone, pp 831-855.
27. Byzova TV, Plow EF: Networking in the hemostatic system: 46. de Alarcon PA, Graeve JA, Levine RF, et al: Thrombocytopenia
integrin IIb3 binds prothrombin and influences its activa- and absent radii syndrome: defective megakaryocytopoiesis-
tion. J Biol Chem 272:27183-27188, 1997. thrombocytopoiesis. Am J Pediatr Hematol Oncol 13:77-83,
28. Gemmell CH, Sefton MV, Yeo EL: Platelet-derived micro 1991.
particle formation involves glycoprotein IIb-IIIa: inhibition 47. Raccuglia G: Gray platelet syndrome: a variety of qualitative
by RGDs and a Glanzmanns thrombasthenia defect. J Biol platelet disorder. Am J Med 51:818-828, 1971.
Chem 268:14586-14589, 1993. 48. Berrebi A, Klepfish A, Varon D, et al: Gray platelet syndrome
29. Reverter JC, Beguin S, Kessels H, et al: Inhibition of platelet- in the elderly. Am J Hematol 28:270-272, 1988.
mediated tissue factor-induced thrombin generation by the 49. Pfueller SL, Howard MA, White JG, et al: Shortening of the
mouse/human chimeric 7E3 antibody: potential implica- bleeding time by 1-deamino-8-arginine vasopressin (DDAVP)
tions for the effect of c7E3 Fab treatment on acute thrombosis in the absence of platelet von Willebrand factor in gray plate-
and clinical restenosis. J Clin Invest 98:863-874, 1996. let syndrome. Thromb Haemost 58:1060-1063, 1987.
CHAPTER 44 Qualitative Disorders of Platelets and Vasculature 733
50. Brace LD, Venton DL, Le Breton GC: Thromboxane A2/ 71. Bick RL, Scates SM: Qualitative platelet defects. Lab Med
prostaglandin H2 mobilizes calcium in human blood plate- 23:95-103, 1992.
lets. Am J Physiol 249:H1-H7, 1985. 72. Haut MJ, Cowan DH: The effect of ethanol on hemostatic
51. Moake JL, Funicella T: Common bleeding problems. Clin properties of human blood platelets. Am J Med 56:22-33,
Symp 35:1-32, 1983. 1974.
52. Nieuwenhuis HK, Sakariassen KS, Houdijk WPM, et al: Defi- 73. Wilhite DB, Comerota AJ, Schmieder FA, et al: Managing PAD
ciency of platelet membrane glycoprotein Ia associated with with multiple platelet inhibitors: the effect of combination
a decreased platelet adhesion to subendothelium: a defect in therapy on bleeding time. J Vasc Surg 38:710-713, 2003.
platelet spreading. Blood 68:692-695, 1986. 74. Lopez J, Thiagarajan P: Acquired disorders of platelet func-
53. Moroi M, Jung SM, Okuma M, et al: A patient with platelets tion. In Hoffman R, Benz EJ, Jr, Shattil SJ, editors: Hematology:
deficient in glycoprotein VI that lack both collagen-induced basic principles and practice, ed 4, Philadelphia, 2005, Churchill
aggregation and adhesion. J Clin Invest 84:1440-1445, 1989. Livingstone, pp 2347-2367.
54. Nurden P, Jandrot-Perrus M, Combrie R, et al: Severe defi- 75. Schleinitz MD, Heidenreich PA: A cost-effectiveness analysis
ciency of glycoprotein VI in a patient with gray platelet syn- of combination antiplatelet therapy of high-risk acute coro-
drome. Blood 104:107-114, 2004. nary syndromes: clopidogrel plus aspirin versus aspirin alone.
55. Cattaneo M: Inherited platelet-based bleeding disorders. Ann Intern Med 142:251-259, 2005.
J Thromb Haemost 1:1628-1636, 2003. 76. Coller BS: GPIIb/IIIa antagonists: pathophysiologic and ther-
56. Daniel JL, Dangelmaier C, Jin J, et al: Molecular basis for apeutic insights from studies of c7E3 Fab. Thromb Haemost
ADP-induced platelet activation: I. Evidence for three distinct 78:730-735, 1997.
ADP receptors on human platelets. J Biol Chem 273:2024- 77. Tcheng JE: Platelet glycoprotein IIb/IIIa integrin blockade:
2029, 1998. recent clinical trials in interventional cardiology. Thromb
57. Nurden P, Savi P, Heilmann E, et al: An inherited bleeding Haemost 78:205-209, 1997.
disorder linked to a defective interaction between ADP and 78. Van de Werf F: Clinical trials with glycoprotein IIb/IIIa recep-
its receptor on platelets. J Clin Invest 95:1612-1622, 1995. tor antagonists in acute coronary syndromes. Thromb Haemost
58. Cattaneo M, Zighetti ML, Lombardi R, et al: Molecular basis 78:210-213, 1997.
of defective signal transduction in the platelet P2Y12 receptor 79. Landolfi R, Marchioli R, Patrono C: Mechanisms of bleeding
of a patient with congenital bleeding. Proc Natl Acad Sci and thrombosis in myeloproliferative disorders. Thromb
U S A 100:1978-1983, 2003. Haemost 78:617-621, 1997.
59. Rao AK: Congenital disorders of platelet function: disorders 80. Swart SS, Pearson D, Wood JK, et al: Functional significance
of signal transduction and secretion. Am J Med Sci 316:69-76, of the platelet alpha2-adrenoreceptor: studies in patients
1998. with myeloproliferative disorders. Thromb Res 33:531-541,
60. Rao AK: Inherited defects in platelet signaling mechanisms. 1984.
J Thromb Haemost 1:671-681, 2003. 81. Bosch J, Thabut D, Bendtsen F, et al: Recombinant factor VIIa
61. Zhou Q, Sims PJ, Wiedmer T: Expression of proteins control- for upper gastrointestinal bleeding in patients with cirrhosis:
ling transbilayer movement of plasma membrane phospho- a randomized, double-blind trial. Gastroenterology 127:1123-
lipids in B lymphocytes from a patient with Scott syndrome. 1130, 2004.
Blood 92:1707-1712, 1998. 82. Boccardo P, Remuzzi G, Galbusera M: Platelet dysfunction in
62. Zhou Q, Zhao J, Stout JG, et al: Molecular cloning of human renal failure. Semin Thromb Hemost 30:579-589, 2004.
plasma membrane phospholipid scramblase: a protein medi- 83. Martinez J, Ferber A: Disorders of fibrinogen. In Hoffman R,
ating transbilayer movement of plasma membrane phospho- Benz EJ, Jr, Shattil SJ, et al, editors: Hematology: basic principles
lipids. J Biol Chem 272:18240-18244, 1997. and practice, ed 4, Philadelphia, 2005, Churchill Livingstone,
63. Zwaal RF, Comfurius P, Bevers EM: Scott syndrome, a bleed- pp 2097-2109.
ing disorder caused by a defective scrambling of membrane 84. Eldrup-Jorgensen J, Flanigan DP, Brace LD, et al: Hypercoagu-
phospholipids. Biochim Biophys Acta 1636:119-128, 2004. lable states and lower limb ischemia in young adults. J Vasc
64. Sims P, Wiedmer T: Unraveling the mysteries of phospholipid Surg 9:334-341, 1989.
scrambling. Thromb Haemost 86:266-275, 2001. 85. Helgason CM, Hoff JA, Kondos GT, et al: Platelet aggregation
65. Stormorken H, Holmsen H, Sund R, et al: Studies on the in patients with atrial fibrillation taking aspirin or warfarin.
haemostatic defect in a complicated syndrome: an inverse Stroke 24:1458-1461, 1993.
Scott syndrome platelet membrane abnormality? Thromb 86. Pareti FI, Capitanio A, Mannucci L, et al: Acquired dysfunc-
Haemost 74:1244-1251, 1995. tion due to circulation of exhausted platelets. Am J Med
66. Helgason CM, Bolin KM, Hoff JA, et al: Development of 69:235-240, 1980.
aspirin resistance in persons with previous ischemic stroke. 87. Coller BS, Schneiderman PI: Clinical evaluation of hemor-
Stroke 25:2331-2336, 1994. rhagic disorders: the bleeding history and differential diag-
67. Helgason CM, Tortorice KL, Winkler SR, et al: Aspirin response nosis of purpura. In Hoffman R, Benz EJ, Jr, Shattil SJ,
and failure in cerebral infarction. Stroke 24:345-350, 1993. et al, editors: Hematology: basic principles and practice, ed 4,
68. Beardsley DS: Hemostasis in the perinatal period: approach Philadelphia, 2005, Churchill Livingstone, pp 1824-1840.
to the diagnosis of coagulation disorders. Semin Perinatol 88. Maceyko RF, Camisa C: Kasabach-Merritt syndrome. Pediatr
15(suppl 2):25-34, 1991. Dermatol 8:133-136, 1991.
69. Eikelboom JW, Hirsch J, Weitz J, et al: Aspirin-resistant 89. Brace LD: The multiple myelomas: a review of selected
thromboxane biosynthesis and the risk of myocardial infarc- aspects. Allied Health Behav Sci 3:47-61, 1981.
tion, stroke, or cardiovascular death in patients at high risk 90. Gertz MA, Lacy MQ, Dispenzieri A: Amyloidosis. In Hoffman
for cardiovascular events. Circulation 105:1650-1655, 2002. R, Benz EJ, Jr, Shattil SJ, et al, editors: Hematology: basic
70. Green D: Diagnosis and management of bleeding disorders. principles and practice, ed 4, Philadelphia, 2005, Churchill
Compr Ther 14:31-36, 1988. Livingstone, pp 1537-1560.
45 Laboratory Evaluation of
Hemostasis
George A. Fritsma

OUTLINE OBJECTIVES
Hemostasis Specimen After completion of this chapter, the reader will be able to:
Collection
Patient Management 1. Properly collect and transport hemostasis blood 9. Interpret clot-based screening test results collec-
During Hemostasis specimens. tively to reach presumptive diagnoses, then recom-
Specimen Collection 2. Reject hemostasis blood specimens due to clots, mend and perform confirmatory tests.
Hemostasis Specimen short draws, or hemolysis. 10. Perform partial thromboplastin time mixing studies
Collection Tubes 3. Prepare hemostasis blood specimens for analysis. to detect factor deficiencies, lupus anticoagulants,
Hemostasis Specimen 4. Describe the principles of platelet aggregometry. and specific factor inhibitors.
Collection Rules 5. Apply appropriate platelet function tests in a variety 11. Describe the principle of, appropriately select, and
Specimen Collection of conditions and interpret their results. correctly interpret coagulation factor assays.
Using Syringes and 6. Diagnose von Willebrand disease and monitor its 12. Describe the principle of and correctly interpret
Winged-Needle Sets
treatment. Bethesda titers for coagulation factor inhibitors.
Selection of Needles for
7. Analyze plasma markers of platelet activation platelet 13. Describe the principle of, appropriately select, and
Hemostasis Specimens
Collection of Specimens factor 4 and -thromboglobulin. correctly interpret tests of fibrinolysis, including
from Vascular Access 8. Describe the principle of, appropriately select, and assays for D-dimer, plasminogen, plasminogen acti-
Devices correctly interpret the results of clot-based coagula- vators, and plasminogen activator inhibitors.
Specimen Collection tion screening tests, including activated clotting
Using Capillary Puncture time, prothrombin time, partial thromboplastin time,
Anticoagulants Used for and the thrombin clotting time.
Hemostasis Specimens
Hemostasis Specimen
Management
Hemostasis Specimen
Storage Temperature
Hemostasis Specimen
Storage Time
Preparation of Hemostasis CASE STUDY
Specimens for Assay After studying the material in this chapter, the reader should be able to respond to the following
Platelet Function Tests case study:
Bleeding Time Test for
Platelet Function A 54-year-old woman experienced a pulmonary embolism on September 26 and began oral anti-
Platelet Aggregometry coagulant therapy. Monthly PT values were collected to monitor therapy. From October through
and Lumiaggregometry January, her INR was stable at 2.4, but on February 1 her INR was 1.3. The reduced INR was reported
Testing for Heparin-Induced to her physician.
Thrombocytopenia with On questioning, the patient reported that there had been no change in her warfarin (Coumadin)
Thrombosis dosage or in her diet. She recalled, however, that the phlebotomist had used a tube with a red and
Quantitative Measurement
black stopper. She had thought this to be out of the ordinary and had remarked about it to the
of Platelet Markers
phlebotomist, who made no response. The medical laboratory scientist who had performed the PT
Immunoassay for the
AntiPlatelet Factor 4 assay reexamined the blood specimen and saw that it was in a blue-topped tube.
(Heparin-Induced 1. What did the phlebotomist do?
Thrombocytopenia) 2. What was the consequence of this action?
Antibody 3. What else could cause an unexpectedly short PT?

734
CHAPTER 45 Laboratory Evaluation of Hemostasis 735

OUTLINEcontd HEMOSTASIS SPECIMEN COLLECTION


Assays for Platelet Most hemostasis laboratory procedures are performed on venous whole blood collected by venipunc-
Activation Markers ture and mixed 9:1 with a 3.2% solution of sodium citrate anticoagulant. The specimen is maintained
Clot-Based Plasma as well-mixed whole blood for platelet function testing or centrifuged to provide platelet-poor plasma
Procoagulant Screens (PPP) for other procedures. Phlebotomists, patient care technicians, nurses, medical laboratory scien-
Prothrombin Time
tists, and other health personnel who collect blood specimens must adhere closely to published
Partial Thromboplastin
protocols for specimen collection and management. The nursing or laboratory supervisor is respon-
Time
Partial Thromboplastin sible for the current validity of specimen collection and handling protocols and ensures that personnel
Time Mixing Studies employ approved techniques.1
Thrombin Clotting Time
Reptilase Time Patient Management During Hemostasis Specimen Collection
Coagulation Factor Patients need not fast for hemostasis testing, and little special preparation is required before the
Assays collection of hemostasis laboratory specimens. However, there are numerous drugs that affect the
Fibrinogen Assay outcomes of coagulation tests; for example, aspirin suppresses most platelet function and warfarin
Single-Factor Assays (Coumadin) reduces the activities of factor II (prothrombin), factor VII, factor IX, factor X, protein C,
Using the Partial protein S, and protein Z and prolongs the prothrombin time (PT). The phlebotomist should attempt
Thromboplastin
to record all drugs the patient is currently taking, and patients should be instructed by their physicians
Time Test
to discontinue drugs that may interfere with coagulation test results before testing.
Bethesda Titer for
AntiFactor VIII Inhibitor Phlebotomists may manage patients using standard protocols for identification, cleansing, tourni-
Single-Factor Assays quet use, and venipuncture (see Chapter 3). If there is a reason to anticipate excessive bleedingfor
Using the Prothrombin instance, if the patient has multiple bruises or mentions a tendency to bleedthe phlebotomist should
Time Test extend the time for observing the venipuncture site from 1 minute to 5 minutes and should apply a
Factor XIII Assay: 5-mol/L pressure bandage before dismissing the patient.
Urea Solubility
Tests of Fibrinolysis Hemostasis Specimen Collection Tubes
Quantitative D-Dimer Most hemostasis specimens are collected in plastic blue-stopper (blue-top, blue-closure) sterile evacuated
Immunoassay blood collection tubes containing a measured volume of 0.105 to 0.109mol/L (3.2%) buffered sodium
Fibrin Degradation
citrate anticoagulant.2 Tubes of uncoated soda-lime glass are unsuitable because their negative surface
Product Immunoassay
charge activates platelets and plasma procoagulants. Siliconized (plastic-coated) glass tubes are avail-
Plasminogen
Chromogenic Substrate able, but their use is waning because of concern for potential breakage with consequent risk of exposure
Assay to bloodborne pathogens.3
Tissue Plasminogen
Activator Assay Hemostasis Specimen Collection Rules
Plasminogen Activator If the hemostasis specimen is part of a series of tubes to be filled from a single venipuncture site, it must
Inhibitor 1 Assay be collected first or immediately after a nonadditive tube. The hemostasis tube may not immediately
Euglobulin Clot Lysis follow a tube that contains heparin (green stopper), ethylenediamine tetraacetic acid (EDTA, lavender
Time Test stopper), sodium fluoride (gray stopper), or clot-promoting silica particles such as are contained in
Global Coagulation Assay: plastic red-topped or serum separator (gel) tubes. These additives may become transferred to the
The Thromboelastograph
hemostasis specimen on the stopper needle and invalidate all hemostasis test results. Nonadditive
tubes includes red-topped glass tubes and clear-topped or red and gray marbletopped tubes. If nonad-
ditive tubes are unavailable, the phlebotomist may use and discard a preliminary blue-topped tube.4
The ratio of whole blood to anticoagulant must be 9 parts blood to 1 part anticoagulant. Evacuated
tubes are designed so that the negative internal pressure draws the correct volume of blood from
the vein. Collection tube manufacturers indicate the allowable range of collection volume error in
package inserts and provide a minimum volume line on each tube. In most cases, the volume of
blood collected must be within 90% of the calibrated volume. A short drawthat is, a specimen
with a smaller volume than that specified by the manufacturergenerates erroneously prolonged
clot-based coagulation test results because the excess anticoagulant relative to blood volume neu-
tralizes test reagent calcium.5 Short-draw specimens are consistently discarded, and a fresh specimen
is collected from the patient. Most plastic blue-topped tubes collect 3.0mL of whole blood; the
smaller the collection tube, the narrower the tolerance for short draws.
When specimens are collected using winged-needle butterfly sets, the phlebotomist must compensate
for the internal volume of the tubing, which is usually 12 inches long and contains approximately
0.5mL of air. The phlebotomist must first collect and discard a nonadditive tube or an identical
blue-topped tube. This step ensures that the needle set tubing is filled with fresh patient blood
before the hemostasis specimen is collected.6
736 PART VIII Hemostasis and Thrombosis

TABLE 45-1 Hemostasis Specimen Collection Errors That Require Collection of a New Specimen
Error Comments
Short draw Whole-blood volume less than 90% of required volume or less than manufacturer-specified minimum
Clot in specimen Each specimen must be visually inspected prior to centrifugation; the presence of even a small clot
requires that the specimen be recollected.
Visible hemolysis Hemolysis, pink or red plasma, indicates in vitro activation of platelets and coagulation, and results
will be unreliable.
Lipemia or icterus Optical instruments may not measure clots in cloudy or highly colored specimens. The scientist
automatically shifts to the use of a mechanical instrument.
Prolonged tourniquet application Stasis elevates the concentration of von Willebrand factor and factor VIII and shortens the measured
time on clot-based tests.
Specimen storage at 1 C to 6 C Storage at refrigerator temperatures causes precipitation of large von Willebrand factor multimers
and destroys platelet integrity.
Specimen storage at more than 25 C Storage at above standard room temperature causes coagulation factor VIII deterioration.

Clotted specimens are useless for hemostasis testing, even if blood collection units. Syringes, joined with winged-needle
the clot is small. A few seconds after collection the phle- sets, are especially useful for collecting specimens from patients
botomist must gently invert the specimen at least five times whose veins are small, fragile, or scarred by repeated venipunc-
to mix the blood with the anticoagulant and prevent clot tures. The use of syringes presents additional needle stick risk
formation. If possible, the medical laboratory scientist must to the phlebotomist, so careful training and handling are
visually examine for clots just before centrifugation and essential.10
testing. Many coagulometers are equipped to detect the The phlebotomist selects sterile syringes of 20-mL capacity
presence of clots. Clotted specimens are discarded, and a or less with nonthreaded Luer-slip hubs. The phlebotomist
new specimen is collected from the patient. assembles syringes, a winged needle set (Figure 45-1), a tubing
Excessive specimen agitation causes hemolysis, procoagu- clamp, and standard venipuncture materials. The phlebotomist
lant activation, and platelet activation. The phlebotomist then uses the following protocol:
must never shake the tube. The test results from visibly hemo- 1. Use standard patient identification and standard blood
lyzed specimens are unreliable, and the specimen must be specimen management precautions (see Chapter 3)
recollected (Table 45-1).7 2. Most syringes are delivered with the plunger withdrawn
Excess manipulation of the needle may promote the release about 1mm from the end of the barrel. Move the plunger
of procoagulant substances from the skin and connective outward and inward within the barrel. Expel all air from the
tissue, which contaminate the specimen and cause clotting barrel and affix the needle set to the Luer-slip hub.
factor activation. Consequently, test results from specimens 3. Optional: draw precisely measured anticoagulant into the
collected during a traumatic venipuncture may be falsely syringe prior to collection.
shortened and unreliable.8 4. Cleanse the venipuncture site, affix the tourniquet, and
During blood collection, the phlebotomist must remove the insert the winged needle. Immobilize the needle set by
tourniquet within 1 minute of its application to avoid loosely taping the tube to the arm about 2 inches from the
stasis.9 Stasis is a condition in which venous flow is slowed. needle.
Stasis results in the local accumulation of coagulation factor 5. Fill the syringe using a gentle, even pressure.
VIII and von Willebrand factor (VWF), which may result in 6. Place the syringe on a clean surface and clamp the tubing
false shortening of clot-based coagulation test results. with a hemostat near the needle hub.
Managers of many hemostasis specialty laboratories insist 7. Attach a second syringe if needed; release the clamp and fill
that specimens from patients from whom it is difficult to draw the second syringe.
blood and specimens for specialized tests such as platelet 8. Replace the clamp, remove the needle set, and immediately
aggregometry be collected by syringe, as described in the fol- activate the needle cover.
lowing sections. Many hemostasis laboratories employ medical After seeing to the patients welfare, the phlebotomist cau-
laboratory scientists and phlebotomists who are specially tiously transfers the blood specimen to sealed evacuated tubes
trained in specimen collection to ensure the integrity of the by affixing a safety transfer device. The specimen is allowed to
specimen. flow gently down the side of the tube. The specimen is not
pushed forcibly into the tube, because agitation causes hemo-
Specimen Collection Using Syringes lysis and platelet activation. The transfer must be accomplished
and Winged-Needle Sets within a few seconds of the time the syringe is filled, and the
Although syringes are impractical for high-volume hemostasis tube must be gently inverted at least five times. The specimen
screening, they are occasionally used in place of evacuated volume must be correct.
CHAPTER 45 Laboratory Evaluation of Hemostasis 737

10-mL plastic syringe

Luer-slip hub

Syringe with anticoagulant

1 mL 3.2% sodium citrate

12-inch tubing

Luer-slip hub

"Wings"

A 1-inch needle

B
Figure 45-1 A, Winged needle set and syringe for collecting special hemostasis specimens. The phlebotomist may use the option of drawing the desired
volume of anticoagulant into the syringe prior to blood collection. B, Winged needle illustrating needle-covering safety interlock.

Selection of Needles for overall specimen is 25mL, a 20- or 21-gauge thin-walled needle
Hemostasis Specimens is used (Table 45-2). For a larger specimen, a 19-gauge needle
Whether evacuated collection tubes or syringes are used, the is required. A 23-gauge needle is acceptable for pediatric
bore of the needle should be sufficient to prevent hemolysis patients or patients whose veins are small, but the negative
and activation of platelets and plasma procoagulants. If the collection pressure must be reduced. All needles provide safety
738 PART VIII Hemostasis and Thrombosis

TABLE 45-2 Selection of Needles for Hemostasis that is just off center of the fingertip and perpendicular to the
Specimens fingerprint lines. After wiping away the first drop of blood,
which is likely to be contaminated by tissue fluid, the phle-
Preferred Needle Gauge
botomist places the collection device directly adjacent to the
Application and Length
free-flowing blood and allows the device to fill. The phleboto-
Adult with good veins, 20 or 21 gauge, thin walled, 1.0 or mist wipes excess blood from the outside of the device and
specimen 25mL 1.25 inches long introduces it to the coagulometer to complete the assay. The
Adult with good veins, 19 gauge, 1.0 or 1.25 inches long phlebotomist then presses a gauze pad to the wound and
specimen 25mL instructs the patient to maintain pressure until bleeding ceases.
Child or adult with small, 23 gauge, winged-needle set; apply The key to accurate measurement of PT is a free-flowing
friable, or hardened veins minimal negative pressure puncture. Often it is necessary for the phlebotomist to warm
Transfer of blood from syringe 19 gauge the patients hand to increase blood flow to the fingertips.
to tube Blood collection device distributors provide dry disposable
Syringe with winged-needle set 20 or 21 gauge, thin walled; use only warming devices for this purpose. The phlebotomist avoids
for small, friable, or hardened veins squeezing (milking) the finger, because this renders the
or special coagulation testing blood specimen inaccurate by raising the concentration of
tissue fluid relative to blood cells.14

Anticoagulants Used for


closures that either cover or blunt the needle immediately after Hemostasis Specimens
completion of the venipuncture. Sodium Citrate (Primary Hemostasis Anticoagulant)
The anticoagulant used for hemostasis testing is buffered
Collection of Specimens from Vascular 3.2% (0.105 to 0.109mol/L) sodium citrate, Na3C6H5O72H2O,
Access Devices molecular weight 294.10. Sodium citrate binds calcium ions to
Blood specimens may be drawn from heparin or saline prevent coagulation, and the buffer stabilizes specimen pH as
locks, ports in intravenous lines, peripherally inserted central long as the tube stopper remains in place.15
catheters (PICC tubes), central venous catheters, or dialysis The anticoagulant solution is mixed with blood to produce
catheters. Vascular access device management requires strict a 9:1 ratio; 9 parts whole blood to 1 part anticoagulant. In
adherence to protocol to ensure sterility, prevent emboli, and most cases, 0.3mL of anticoagulant is mixed with 2.7mL of
prevent damage to the device. Personnel must be trained and whole blood, which are the volumes in the most commonly
must recognize the signs of complications and take appropriate used evacuated plastic collection tubes, but any volumes are
action. Institutional protocol may limit vascular access device valid, provided that the 9:1 ratio is maintained. The ratio
blood collection to physicians and nurses. Before blood is col- yields a final citrate concentration of 10.5 to 10.9mmol/L of
lected for hemostasis testing, the line must be flushed with anticoagulant in whole blood.16 Some laboratory scientists
5mL of saline, and the first 5mL of blood, or six times the prepare specimen tubes locally for special hemostasis testing.
volume of the tube, must be collected and discarded. The phle-
botomist must not flush with heparin. Blood is collected into a Adjustment of Sodium Citrate Volume for Elevated
syringe and transferred to an evacuated tube as described in the Hematocrits. The 9:1 blood-to-anticoagulant ratio is effec-
prior section on hemostasis specimen collection with syringes tive provided the patients hematocrit is 55% or less. In poly-
and winged needle sets.11 cythemia, the decrease in plasma volume relative to whole
blood unacceptably raises the anticoagulant-to-plasma ratio,
Specimen Collection Using which causes falsely prolonged results on clot-based coagula-
Capillary Puncture tion tests. The phlebotomist must provide tubes with relatively
Several near-patient testing (point-of-care) coagulometers (see reduced anticoagulant volumes for collection of blood from a
Chapter 47) generate PT results from a specimen consisting of patient whose hematocrit is known to be 55% or higher. The
10 to 50mcL of whole blood. These instruments are designed amount of anticoagulant needed may be computed for a 5-mL
to test either anticoagulated venous whole blood or capillary total specimen volume by using the graph in Figure 45-2 or the
(finger-stick) blood and represent a significant convenience to following formula, which is valid for any total volume:
patients and to anticoagulation clinics.12 Many are designed for
patient self-testing, and pediatric or neonatal testing, and labo- C = (1.85 103 ) (100 H) V
ratory scientists are often charged with training patients in
proper capillary puncture technique.13 where C is the volume of sodium citrate in milliliters, V is
Capillary specimen punctures are made using sterile spring- volume of whole bloodsodium citrate solution in milliliters,
loaded lancets designed to make a cut of standard depth and and H is the hematocrit in percent.
width while avoiding injury (see Chapter 3). The phlebotomist For example, to collect 3mL of blood and anticoagulant
or patient selects and cleanses the middle or fourth (ring) mixture from a patient who has a hematocrit of 65%, calculate
finger and activates the device so that it produces a puncture the volume of sodium citrate as follows:
CHAPTER 45 Laboratory Evaluation of Hemostasis 739

0.5

0.45

0.4

mL Anticoagulant
0.35

0.3

0.25

0.2

0.15
50 55 60 65 70 75 80
Hematocrit
Figure 45-2 Graph for computing the volume of anticoagulant in a 5-mL specimen when the patients hematocrit is 55% or greater. (From Ingram GIC,
Brozovic M, Slater NGP: Bleeding disorders, investigations, and management, ed 2, Oxford, 1982, Blackwell, pp 244-245.)

C = (1.85 103 ) (100 65% ) 3.0 mL citrate dextrose (ACD, yellow stopper) and dipotassium EDTA
C = (1.85 103 ) (35% ) 3.0 mL (K2EDTA) with gel (white stopper) tubes may be used for
C = 0.19 mL of 3.2% sodium citrate molecular diagnosis, as specified by institutional protocol.
Heparinized specimens have never been validated for use in
Remove the tube stopper and pipette and discard 0.11mL plasma coagulation testing but may be necessary in cases of
from the 0.3mL of anticoagulant, leaving 0.19mL. Collect platelet satellitosis (satellitism) as a substitute for specimens
blood in a syringe and transfer approximately 2.89 (2.9) mL collected in EDTA or sodium citrate. Citrate theophylline adenos-
of blood to the tube; replace the stopper, and immediately mix ine dipyridamole (CTAD, blue stopper) tubes are used to halt in
by inverting four times. Some highly skilled laboratory scien- vitro platelet or coagulation activation for specialty assays such
tists actually puncture the stopper with a tuberculin syringe as those for the platelet activation markers platelet factor 4 (PF4)
equipped with a 25-gauge needle, invert the tube, and with- and platelet surface membrane P-selectin (measured by flow
draw the computed volume, leaving the negative pressure cytometry) or the coagulation activation markers prothrombin
intact. Alternatively, the laboratory scientist can prepare for fragment 1+2 and thrombin-antithrombin.
collection of 10mL of blood and anticoagulant solution in a
12-mL centrifuge tube as follows:
HEMOSTASIS SPECIMEN MANAGEMENT
C = (1.85 10 ) (35% ) 10.0 mL
3
Hemostasis Specimen Storage Temperature
C = 0.64 mL of 3.2% sodium citrate Sodium citrateanticoagulated whole-blood specimens are
placed in a rack and allowed to stand in a vertical position with
In this instance, 0.64mL of sodium citrate is pipetted into the stopper intact and uppermost. The pH remains constant as
the tube and 9.36 (9.4) mL of whole blood is transferred from long as the specimen is sealed. Specimens are maintained at
the collection syringe. 18 C to 24 C (room temperature), never at refrigerator tem-
There is no evidence suggesting a need for increasing the peratures (Table 45-3). Storage at 1 C to 6 C activates factor
volume of anticoagulant for specimens from patients with VII, activates platelets, and causes the cryoprecipitation of large
anemia, even when the hematocrit is less than 20%. VWF multimers.18,19 Also, specimens should never be stored at
temperatures greater than 24 C, because heat causes deteriora-
Other Anticoagulants Used for tion of coagulation factor VIII.
Hemostasis Specimens
Few data support the use of EDTA-anticoagulated specimens Hemostasis Specimen Storage Time
for coagulation testing, because calcium ion chelation inter- Specimens collected for PT testing may be held at 18 C to
feres with coagulation assays.17 EDTA is the anticoagulant used 24 C and tested within 24 hours of the time of collection.
in collecting specimens for complete blood counts, including Specimens collected for partial thromboplastin time (PTT)
platelet counts. EDTA may be required for specimens used for testing also may be held at 18 C to 24 C, but must be tested
molecular diagnostic testing, such as testing for factor V Leiden within 4 hours of the time of collection, provided that the speci-
mutation or the prothrombin G20210A mutation. Likewise, acid men does not contain unfractionated heparin anticoagulant. If
740 PART VIII Hemostasis and Thrombosis

TABLE 45-3 Hemostasis Specimen Storage Times and Temperatures


Application Temperature Time
PT with no unfractionated heparin present in specimen 18-24 C 24hr
PTT with no unfractionated heparin present in specimen 18-24 C 4hr
PTT for monitoring unfractionated heparin therapy 18-24 C Separate within 1hr, test within 4hr
PT when unfractionated heparin is present in specimen 18-24 C Separate within 1hr, test within 4hr
Factor assays 18-24 C 4hr
Optical platelet aggregometry using platelet-rich plasma 18-24 C Wait 30min after centrifugation, test within 3hr of collection
Whole-blood aggregometry 18-24 C Test within 3hr of collection
Storage in household freezer 20 C 2wk
Storage for 6mo 70 C 6mo

PT, Prothrombin time; PTT, partial thromboplastin time.

a patient is receiving unfractionated heparin therapy, speci- citrateanticoagulated whole blood is centrifuged at 1500xg
mens for PTT testing must be centrifuged within 1 hour of the for 15 minutes in a swinging bucket centrifuge to produce
time of collection, and the plasma, which should be PPP, must supernatant PPP. Alternatively, the angle-head StatSpin Express
tested within 4 hours of the time of collection.20 2 (Iris Sample Processing, Inc., Westwood, Mass.) generates
4400xg and can produce PPP within 3 minutes. Both make it
Preparation of Hemostasis Specimens possible for automated coagulometers to sample from the
for Assay supernatant plasma of the primary tube. The advantage of
Whole-Blood Specimens Used for the slower swinging bucket centrifuge head is that it produces
Platelet Aggregometry a straight, level plasmablood cell interface, whereas angle-
Blood for whole-blood platelet aggregometry or lumiag- head centrifuge heads cause platelets to adhere to the side
gregometry must be collected with 3.2% sodium citrate and of the tube. If the tube is allowed to stand, the platelets
held at 18 C to 24 C until testing. Chilling destroys platelet drift back into the plasma and release granule contents. Each
activity. Aggregometry may be started immediately and must hemostasis laboratory manager establishes the correct centrifu-
be completed within 3 hours of specimen collection. The sci- gation speed and times for the local laboratory, and centrifu
entist mixes the specimen by gentle inversion, checks for clots gation must yield PPP from specimens with high initial
just before testing, and rejects specimens with clots. Most spe platelet counts.
cimens for whole-blood aggregometry are mixed 1:1 with In the special hemostasis laboratory the manager may
normal saline before testing, although if the platelet count is choose a double-spin approach. The primary tube is centrifuged
less than 100,000/mcL the specimen is tested undiluted.21 using a swinging bucket centrifuge, and the plasma is trans-
ferred to a secondary tube, which is labeled and centrifuged
Platelet-Rich Plasma Specimens Used for again. The double-spin approach may be used to produce PPP
Platelet Aggregometry with a plasma platelet count of less than 5000/mcL, which is
Light-transmittance (optical) platelet aggregometers are preferred for lupus anticoagulant (LA) testing and for prepara-
designed to test platelet-rich plasma (PRP), plasma with a tion of frozen plasma.
platelet count of 200,000 to 300,000/mcL. Sodium citrate The presence of greater than 10,000 platelets/mcL in plasma
anticoagulated blood is first checked visually for clots and then affects clot-based test results. Platelets are likely to become
centrifuged at 50g for 30 minutes with the stopper in place to activated in vitro and release the membrane phospholipid phos-
maintain the pH. The supernatant PRP is transferred by a phatidylserine, which triggers plasma coagulation and neutral-
plastic pipette to a clean plastic tube, and the tube is sealed izes LA if present, interfering with LA testing. Platelets also
and stored at 18 C to 24 C (room temperature) until the test secrete fibrinogen, factors V and VIII, and VWF (see Chapter
is begun. PRP-based light-transmittance aggregometry is initi- 13). These may desensitize PT and PTT assays and interfere
ated no less than 30 minutes after the specimen is centrifuged with clot-based coagulation assays. In addition, platelets release
and completed within 3 hours of the time of collection. To PF4, a protein that binds and neutralizes therapeutic heparin
produce sufficient PRP, the original specimen must measure 9 in vitro, falsely shortening the PTT and interfering with heparin
to 12mL of whole blood. Light-transmittance aggregometry is management.
unreliable when the patients whole-blood platelet count is less The hemostasis laboratory manager arranges to perform
than 100,000/mcL. plasma platelet counts on coagulation plasmas at regular inter-
vals to ensure that they are consistently platelet poor. Many
Platelet-Poor Plasma Required for managers select 10 to 12 specimens from each centrifuge every
Clot-Based Testing 6 months, perform platelet counts, and document that their
Clot-based plasma coagulation tests require PPPplasma samples remain appropriately platelet poor, even if the initial
with a platelet count of less than 10,000/mcL.22 Sodium platelet count is elevated.
CHAPTER 45 Laboratory Evaluation of Hemostasis 741

Laboratory scientists inspect hemostasis plasmas for hemo- thrombocytopenic purpura. Giant or dysplastic platelets are seen
lysis (red), lipemia (cloudy, milky), and icterus (golden yellow in myeloproliferative neoplasms, acute leukemia, and myelo-
from bilirubin). Visible hemolysis implies platelet or coagula- dysplastic syndromes.
tion pathway activation. Visibly hemolyzed specimens are
rejected, and new specimens must be obtained. Lipemia and Bleeding Time Test for Platelet Function
icterus may affect the end-point results of optical coagulation The bleeding time test was the original test of platelet function,
instruments. The hemostasis laboratory manager may choose although it is now largely replaced by near-patient analysis
to maintain a separate mechanical end-point coagulometer to of platelet function using the PFA-100 (Siemens Healthcare
substitute for the optical instrument if the specimen is too Diagnostics, Inc., Deerfield, Ill.), the Ultegra (Accumetrics, San
cloudy for optical determinations. Conversely, some optical Diego, Calif.), or platelet aggregometry.26 To perform the test,
instruments detect and compensate for lipemia and icterus via the phlebotomist uses a lancet to make a small controlled
spectrophotometric analysis.23 puncture wound and records the duration of bleeding, compar-
ing the results with the universally accepted reference interval
Specimen Storage of 2 to 9 minutes. The bleeding time test was first described by
Specimens for PT assay only may be held uncentrifuged at Duke27 in 1912 and modified by Ivy28 in 1941. In 1978 some
18 C to 24 C for up to 24 hours provided the tubes remain standardization was attempted: a blood pressure cuff was
closed. Likewise, specimens for PTT measurement may be held inflated to 40mm Hg, a calibrated spring-loaded lancet
uncentrifuged for up to 4 hours. However, specimens from (Surgicutt Bleeding Time Device; International Technidyne
patients receiving unfractionated heparin collected for PTT Corporation, Edison, N.J.) was triggered on the volar surface
heparin monitoring must be centrifuged and the supernatant of the forearm a few inches distal to the antecubital crease, and
PPP sampled or transferred within 1 hour to avoid false short- the resulting wound was blotted every 30 seconds with filter
ening of the PTT as PF4 neutralizes the heparin. paper until bleeding stopped.29,30
If the hemostasis test cannot be completed within the pre- A prolonged bleeding time could theoretically signal a func-
scribed interval, the laboratory scientist must immediately cen- tional platelet disorder such as von Willebrand disease (VWD)
trifuge the specimen. The supernatant PPP must be transferred or a vascular disorder such as scurvy or vasculitis, and was a
by plastic pipette to a plastic freezer tube, sealed, and frozen, characteristic result of therapy with aspirin and other nonste-
and may be stored at 20 C for up to 2 weeks or at 70 C roidal antiinflammatory drugs (NSAIDs). Measurement of the
for up to 6 months. At the time the test is performed, the speci- bleeding time was often requested by surgeons at admission in
men must be thawed rapidly at 37 C, mixed well, and tested an attempt to predict surgical bleeding, but a series of studies
within 1 hour of the time it is removed from the freezer. If it in the 1990s revealed that the test has inadequate predictive
cannot be tested immediately, the specimen may be stored at value. The bleeding time is affected by the nonplatelet variables
1 C to 6 C for 2 hours after thawing. To avoid cryoprecipita- of intracapillary pressure, skin thickness at the puncture site,
tion of VWF, specimens may not be frozen and thawed more and size and depth of the wound, all of which interfere with
than once. accurate interpretation of the test results. Owing to its poor
predictive value for bleeding and its tendency to scar the
forearm, use of the bleeding time assay has been discontinued
PLATELET FUNCTION TESTS
at most institutions.
Platelet function tests are designed to detect qualitative (func-
tional) platelet abnormalities in patients who are experiencing Platelet Aggregometry
the symptoms of mucocutaneous bleeding (see Chapter 44). A and Lumiaggregometry
platelet count is performed and the blood film is reviewed Functional platelets adhere to subendothelial collagen, aggre-
before platelet function tests are begun, because thrombocyto- gate with each other, and secrete the contents of their -granules
penia is a common cause of hemorrhage (see Chapter 43).24 and -granules (see Chapter 13). Normal adhesion requires
Qualitative platelet abnormalities are suspected only when intact platelet membranes and functional plasma VWF. Normal
bleeding symptoms are present and the platelet count exceeds aggregation requires that platelet membranes and platelet acti-
50,000/mcL. vation pathways are intact, that the plasma fibrinogen concen-
Although hereditary platelet function disorders are rare, tration is normal, and that normal secretions are release from
acquired defects are common.25 Acquired platelet defects are platelet granules. Platelet adhesion, aggregation, and secretion
associated with liver disease, renal disease, myeloproliferative are assessed using in vitro platelet aggregometry.
neoplasms, myelodysplastic syndromes, myeloma, uremia, An aggregometer is an instrument designed to measure
autoimmune disorders, anemias, and drug therapy. Platelet platelet aggregation in a suspension of citrated whole blood or
morphology is often a clue; for instance, in Bernard-Soulier PRP. Specimens are collected and managed in compliance with
syndrome the blood film reveals mild thrombocytopenia and standard laboratory protocol as described in the section on
large gray platelets (see Figure 44-1). Similarly, the presence preparation of hemostasis specimens for assays, and main-
of large platelets on the blood film associated with elevated tained at room temperature (18 C to 24 C) until testing
mean platelet volume often indicates rapid platelet turnover, begins. Specimens for PRP-based light-transmittance aggre
such as occurs in immune thrombocytopenic purpura or thrombotic gometry must stand undisturbed for 30 minutes after
742 PART VIII Hemostasis and Thrombosis

centrifugation while the platelets regain their responsiveness. to move toward 100% light transmission. Platelet deficiencies
Specimens for impedance aggregometry are diluted 1:1 with are reflected in diminished or absent aggregation; many labora-
normal saline and tested immediately. Specimens must be tory directors choose 40% aggregation as the lower limit of
tested within 3 hours of collection to avoid spontaneous normal.
in vitro platelet activation. Platelet aggregometry is a high
complexity laboratory test requiring a skilled, experienced Whole-Blood Platelet Aggregometry
operator. In whole-blood platelet aggregometry, platelet aggregation is
measured by electrical impedance using a 1:1 salinewhole
Platelet Aggregometry Using Platelet-Rich Plasma blood suspension (Model 700 Whole Blood/Optical Lumi-
PRP aggregometry is performed using a specialized photometer Aggregometer; Chrono-log Corporation, Havertown, Pa.).32
called a light-transmittance aggregometer (PAP-8E Platelet Aggre- The operator pipettes aliquots of properly mixed whole blood
gation Profiler; Bio/Data Corporation, Horsham, Pa.).31 After to cuvettes and adds equal volumes of physiologic saline. Sus-
calibrating the instrument in accordance with manufacturer pension volume may be 300 to 500mcL. The operator drops
instructions, the operator pipettes the PRP to instrument- in one stir bar per cuvette and places the cuvettes in 37 C
compatible cuvettes, usually 500 mcL; drops in one clean plas- incubation wells for 5 minutes. The operator transfers the first
ticized stir bar per sample; places the cuvettes in incubation cuvette to a reaction well, forcibly pipettes in an agonist, and
wells; and allows the samples to warm to 37 C for 5 minutes. suspends a pair of low-voltage cartridge-mounted disposable
The operator then transfers the first cuvette, containing speci- direct current (DC) electrodes in the mixture. As aggregation
men and stir bar, to the instruments reaction well and starts occurs, platelets collect on the electrodes, impeding the DC
the stirring device and the recording computer. The stirring current (Figure 45-5). The rise in impedance is directly propor-
device turns the stir bar at 800 to 1200rpm, a gentle speed that tional to platelet aggregation and is amplified and recorded by
keeps the platelets in suspension. The instrument directs instrument circuitry. A whole-blood aggregometry tracing
focused light through the sample cuvette to a photodetector closely resembles a PRP-based light-transmittance aggregome-
(Figure 45-3). As the PRP is stirred, the recorder tracing first try tracing as shown in Figure 45-4.
stabilizes to generate a baseline, near 0% light transmission.
After a few seconds, the operator forcibly pipettes an agonist Platelet Lumiaggregometry
(aggregating agent) into the sample to trigger aggregation. In a The Whole Blood/Optical Lumi-Aggregometer may be used
normal specimen, after the agonist is added, the shape of the for simultaneous measurement of platelet aggregation and
suspended platelets changes from discoid to spherical, and the secretion of adenosine triphosphate (ATP) from activated
the intensity of light transmission increases in proportion to platelet -granules.33 The procedure for lumiaggregometry differs
the degree of shape change. Percent transmittance is monitored little from that for conventional aggregometry and simplifies
continuously and recorded (Figure 45-4). As platelet aggregates the diagnosis of platelet dysfunction.34 As ATP is released it
form, more light passes through the PRP, and the tracing begins oxidizes a firefly-derived luciferin-luciferase reagent (Chrono-
lume; Chrono-log Corporation) to generate cold chemilumi-
nescence proportional to the ATP concentration. A photodetector
Cuvette amplifies the luminescence, which is recorded as a second
tracing on the aggregation report.35
Lumiaggregometry may be performed using whole blood or
Light PRP.36 To perform lumiaggregometry, the operator adds an ATP
source
standard to the first sample, then adds luciferin-luciferase and
Photodetector tests for full luminescence. The operator then adds luciferin-
luciferase and an agonist to the second sample; the instrument
monitors for aggregation and secretion simultaneously. Throm-
bin is typically the first agonist used, because thrombin induces
full secretion. The luminescence induced by thrombin is mea-
sured, recorded, and used for comparison with the lumines-
cence produced by the additional agonists. Normal secretion
induced by agonists other than thrombin produces lumines-
cence at a level of about 50% of that resulting from thrombin.
Platelets in Figure 45-6 depicts simultaneous aggregation and secretion
suspension responses to thrombin; Figure 45-7 is a scanning electron
micrograph of resting and activated platelets.
Stir bar

Figure 45-3 Analysis of platelet-rich plasma in an optical aggregometer. Platelet Agonists (Activating Agents) Used
Desired platelet count is approximately 200,000/mcL. Platelets are main- in Aggregometry
tained in suspension by a magnetic stir bar turning at 800 to 1200rpm. The agonists used most frequently in clinical practice are throm-
(Courtesy Kathy Jacobs, Chrono-log Corporation, Havertown, Pa.) bin or synthetic thrombin receptoractivating peptide (TRAP),
CHAPTER 45 Laboratory Evaluation of Hemostasis 743

Shape
change

Primary Secretion
Resting platelet;
Stable baseline aggregation

0%
10
20
30
% Aggregation

Agonist
40
50
60

Secondary
70

aggregation
80
90
100

0 Time (minutes) 5 minutes


Figure 45-4 Optical aggregometry tracing showing five phases of platelet aggregation: baseline at 0% aggregation, shape change after the addition of
the agonist, primary aggregation, release of adenosine diphosphate and adenosine triphosphate, and second-wave aggregation that forms large clumps.
(Courtesy Kathy Jacobs, Chrono-log Corporation, Havertown, Pa.)

adenosine diphosphate (ADP), epinephrine, collagen, arachi- TABLE 45-4 Platelet Aggregometry Agonists, Reaction
donic acid, and ristocetin. Table 45-4 lists representative con- Concentrations, and Platelet Receptors
centrations and platelet activation pathways tested by each
Typical
agonist. Small volumes (2 to 5mcL) of concentrated agonist
Agonist Concentration Receptors
are used so that they have little dilutional effect in the reaction
system.37 Thrombin 1 unit/mL PAR1 and PAR4; GP Ib
Thrombin (or TRAP) cleaves two platelet membrane and GP V
protease-activatable receptors (PARs), PAR1 and PAR2, both ADP 1-10mcmol/L P2Y1, P2Y12
members of the seven-transmembrane repeat receptor family Epinephrine 2-10mcmol/L 2-Adrenergic receptor
(see Chapter 13). Thrombin or TRAP also cleaves glycoprotein Collagen 5mcg/mL GP Ia/IIa, GP VI
(GP) 1b and GP V. Internal platelet activation is effected Arachidonic acid 500mcmol/L TP, TP
by membrane-associated G proteins and both the eicosanoid Ristocetin 10mg/mL GP Ib/V/IX in association
and the diacylglycerol pathways. Thrombin-induced activation with von Willebrand factor
results in full secretion and aggregation. In lumiaggregometry,
the operator ordinarily begins with 1 unit/mL of thrombin GP, Glycoprotein; PAR, protease-activatable receptor; P2Y, platelet membrane
ADP-receptor; TP, thromboxane receptor.
or TRAP to induce the release of 1 to 2 nmol/L of ATP,
detected by the firefly luciferin-luciferase luminescence assay.
744 PART VIII Hemostasis and Thrombosis

Other agonistsfor instance, 5 mcg/mL of collageninduce


the release of at most 0.5 to 1.0 nmol/L of ATP. Thrombin-
induced secretion may be diminished to less than 1 nmol/L
in storage pool deficiencies (see Chapter 44), but is rela-
tively unaffected by membrane disorders or pathway enzyme
deficiencies.

Cuvette

Electrodes
A

Platelets
aggregate on
electrodes

Platelets in
suspension Stir bar

Figure 45-5 In whole-blood platelet aggregometry, aggregating platelets


form a layer on the electrodes, and current is impeded by the platelet layer. B
Resistance (in ohms) is proportional to aggregation, and a tracing is provided
that resembles the tracing obtained using optical aggregometry. (Courtesy Figure 45-7 Scanning electron micrograph of resting (A) and activated
Kathy Jacobs, Chrono-log Corporation, Havertown, Pa.) (B) platelets.

Impedance ATP release


3.0 mcmol/L
0
Full 1-2 nmol/L
release

20
1.5 mcmol/L

Full 40
aggregation

40
0
0 5 minutes 10 minutes
1 unit/mL thrombin
Figure 45-6 Normal lumiaggregometry tracing illustrating monophasic aggregation curve with superimposed release (secretion) reaction curve. Aggregation
is measured in ohms () using the left y-axis scale; release is measured in mcmol/L of adenosine triphosphate (ATP) based on luminescence using the right
y-axis scale. Curve illustrates full aggregation and secretion response to 1 unit/mL of thrombin. (Courtesy Margaret Fritsma, University of Alabama at
Birmingham.)
CHAPTER 45 Laboratory Evaluation of Hemostasis 745
Reagent thrombin is stored dry at 20 C and is reconsti- Epinephrine is stored at 1 C to 6 C and reconstituted
tuted with physiologic saline immediately before use. Leftover with distilled water immediately before it is used. Leftover
reconstituted thrombin may be divided into aliquots, frozen, reconstituted epinephrine may be aliquoted and frozen for
and thawed for later use. Thrombin has the disadvantage that later use.
it often triggers coagulation simultaneously with aggregation. Collagen binds GP Ia/IIa and GP VI, but induces no primary
The use of TRAP avoids this pitfall. aggregation. After a lag of 30 to 60 seconds, aggregation begins,
ADP binds platelet membrane receptors P2Y1 and P2Y12, and a monophasic curve develops. Aggregation induced by col-
also members of the seven-transmembrane repeat receptor lagen at 5mcg/mL requires intact membrane receptors, func-
family. ADP-induced platelet activation relies on the physio- tional membrane G protein, and normal eicosanoid pathway
logic response of membrane-associated G protein and the eico- function. Loss of collagen-induced aggregation may indicate a
sanoid synthesis pathway. The end product of eicosanoid membrane abnormality, storage pool disorder, release defect,
synthesis, thromboxane A2, raises cytosolic free calcium, which or the presence of aspirin.
mediates platelet activation and induces secretion of ADP Most managers purchase lyophilized fibrillar collagen prep-
stored in -granules. The secreted ADP activates neighboring arations such as Chrono-Par Collagen (Chrono-log Corpo
platelets. ration). Collagen is stored at 1 C to 6 C and used without
ADP is the most commonly used agonist, particularly in further dilution. Collagen may not be frozen.
aggregometry systems that measure only aggregation and not Arachidonic acid assesses the viability of the eicosanoid syn-
luminescence. The operator adjusts the ADP concentration thesis pathway. Free arachidonic acid agonist at 500nmol/L is
to between 1mcmol/L and 10mcmol/L to induce biphasic added to induce a monophasic aggregometry curve with virtu-
aggregation (see Figure 45-4). At ADP concentrations near ally no lag phase. Aggregation is independent of membrane
1mcmol/L, platelets achieve only primary aggregation, followed integrity. Deficiencies in eicosanoid pathway enzymes, includ-
by disaggregation. The recording deflects from the baseline for ing deficient or aspirin-suppressed cyclooxygenase, result in
1 to 2 minutes and then returns to baseline. Primary aggrega- reduced aggregation and secretion.
tion involves shape change with formation of microaggregates, Arachidonic acid is readily oxidized and must be stored at
both reversible. Secondary aggregation is the formation of full 20 C in the dark. The operator dilutes arachidonic acid with
platelet aggregates after release of platelet -granule ADP. At a solution of bovine albumin for immediate use. Aliquots of
agonist ADP concentrations near 10mcmol/L, there is simul- bovine albumindissolved arachidonic acid may be frozen for
taneous irreversible shape change, secretion, and formation of later use.
aggregates, resulting in a monophasic curve and full deflection
of the tracing. ADP concentrations between 1 and 10mcmol/L
induce a biphasic curve: primary aggregation followed by a Platelet Aggregometry Tests
brief flattening of the curve called lag phase and then secondary in von Willebrand Disease
aggregation. Ristocetin-Induced Platelet Aggregation. Although
Operators expend considerable effort to discover the ADP the test is usually called the ristocetin-induced platelet aggregation
concentration that generates a biphasic curve with a visible lag (RIPA) test, ristocetin actually induces an agglutination reaction
phase, because the appropriate concentration varies among with little platelet shape change and little secretion. A normal
patients. This enables operators to use aggregometry alone to RIPA result may imply that normal concentrations of func-
distinguish between membrane-associated platelet defects and tional VWF are present and that the platelets possess a func-
storage pool or release defects. tional VWF receptor, GP Ib/V/IX (see Chapter 13).38
Lumiaggregometry provides a clearer and more reproduc- Ristocetin at 10 mg/mL induces a monophasic aggregation
ible measure of platelet secretion, rendering the quest for the tracing from a normal specimen. Specimens from patients
biphasic curve unnecessary. Secretion in response to ADP at with VWD, except for subtype 2B VWD, produce a reduced
5mcmol/L is diminished in platelet membrane disorders, or absent reaction, although all other agonists generate
eicosanoid synthesis pathway enzyme deficiencies, storage normal tracings (Table 45-5). Exogenous VWF from normal
pool deficiency, or aspirin, NSAID, or clopidogrel therapy. plasma restores a normal RIPA reaction, confirming the diag-
Reagent ADP is stored at 20 C, reconstituted with phy nosis (see Chapter 41). In patients with Bernard-Soulier syn-
siologic saline, and used immediately after reconstitution. drome, a congenital abnormality of the GP Ib or IX portion
Leftover reconstituted ADP may be aliquoted and frozen for of the GP Ib/V/IX receptor results in a diminished RIPA
later use. reaction that is not corrected by the addition of VWF (see
Epinephrine binds platelet -adrenergic receptors, identical Chapter 44).
to muscle receptors, and activates the platelet through the same In subtype 2B VWD, a VWF gain-of-function mutation,
metabolic pathways as reagent ADP. The results of epinephrine- aggregation occurs even when reduced ristocetin concentra-
induced aggregation match those of ADP except that epineph- tions (down to 1mg/mL) are added. This response illustrates
rine cannot induce aggregation in storage pool disorder or the increased affinity of large VWF multimers for platelet
eicosanoid synthesis pathway defects no matter how high its receptors. The low-dose or low-concentration RIPA, sometimes
concentration. Epinephrine does not work in whole-blood called the ristocetin response curve, is used to diagnose type
aggregometry. 2B VWD.
746 PART VIII Hemostasis and Thrombosis

TABLE 45-5 Typical Normal Ranges in Platelet TABLE 45-6 Expected Platelet Aggregometry Results
Aggregometry in Storage Pool Disorder for a Variety of Agonists
Aggregation Aggregation
Agonist Concentration Impedance ATP Secretion Agonist Concentration Impedance ATP Secretion
Thrombin 1 unit Not recorded 1.0-2.0nmol/L Thrombin 1 unit/mL Not recorded <0.1nmol/L
Collagen 1mcg/mL 15-27 0.5-1.7nmol/L Collagen 5mcg/mL 20 <0.1nmol/L
5mcg/mL 15-31 0.9-1.7nmol/L Arachidonic acid 500mcmol/L 12 <0.1nmol/L
ADP 5mcmol/L 1-17 0.0-0.7nmol/L
10mcmol/L 6-24 0.4-1.7nmol/L ATP, Adenosine triphosphate.
Arachidonic acid 500mcmol/L 5-17 0.6-1.4nmol/L
Ristocetin 1mg/mL >10 Not recorded
Therapy with Aspirin, Other Nonsteroidal Antiinflam-
ATP, Adenosine triphosphate; ADP, adenosine diphosphate. matory Drugs, and Clopidogrel. NSAIDs such as aspirin,
ibuprofen, indomethacin, and sulfinpyrazone permanently
inactivate or temporarily inhibit cyclooxygenase. The thieno-
The RIPA test is qualitative and is diagnostic in only about pyridine antiplatelet drugs clopidogrel and prasugrel irreversibly
70% of cases. Most laboratory managers have dropped RIPA occupy the ADP receptor P2Y12. The NSAIDs limit or eliminate
from their test menus because of its poor predictive values. the aggregation and secretion responses to arachidonic acid
There is considerable variation in laboratory results from one and collagen. Thienopyridines suppress aggregation and secre-
patient to another in the same kindred and from time to time tion responses to ADP.41 Platelet aggregometry is employed
in a single patient. Consequently, the laboratory director must to monitor response to these antiplatelet drugs (see Chapter
include the ristocetin cofactor test, the VWF antigen immunoassay, 46).42 The physician or medical laboratory scientist must
and the coagulation factor VIII activity assay in the VWD profile. instruct the patient to discontinue all antiplatelet drugs at
Many laboratories also offer the collagen binding assay. Ultimate least 1 week before blood is collected for aggregometry
confirmation and characterization of VWD is based on gel unless aggregometry is ordered to monitor the effects of
immunoelectrophoresis to characterize VWF monomers (see these drugs.
Chapter 41).39
Platelet Release (Secretion) Defects: Eicosanoid
Quantitative Ristocetin Cofactor Assay for von Wille Pathway Enzyme Deficiencies. Congenital or acquired
brand Disease. One essential refinement of ristocetin deficiencies of cyclooxygenase, thromboxane synthase, protein
aggregometry is the substitution of formalin-fixed or lyophi- kinase C, or any enzyme in the eicosanoid activation pathway
lized normal reagent platelets for the patients platelets.40 limit or prevent secretion. Thrombin induces normal responses,
When reagent platelets are used, the test is called the ristocetin but secretion and aggregation are diminished in response to
cofactor or VWF activity assay. The medical laboratory scientist ADP, epinephrine, collagen, and arachidonic acid. Because the
prepares the patients PPP; mixes it with reagent platelets; adds aggregation responses resemble the responses seen during the
ristocetin; and performs optical, not impedance, aggregometry. use of NSAIDs, release defects are often called aspirin-like
The ristocetin cofactor assay yields a proportional relationship disorders.
between VWF activity and the aggregometry response of the
reagent platelets. Comparison of the aggregation results for Storage Pool Deficiency. In a congenital or acquired
patients PPP with those for standard dilutions of normal PPP storage pool defect, -granules are empty or missing. ATP
permits a quantitative expression of the VWF activity level. The release in response to thrombin is reduced, as it is in
ristocetin cofactor test also is available as an automated assay response to ADP, epinephrine, arachidonic acid, and collagen
on the BCT coagulometer and the BCS coagulometer (Siemens (Table 45-6).
Healthcare Diagnostics), which use latex particles in place of
preserved platelets. Platelet Membrane Defects: Thrombasthenia. Glan-
zmann thrombasthenia, a membrane defect characterized by
Summary of Agonist Responses dysfunction or loss of the GP IIb/IIIa receptor site, may be
in Various Circumstances diagnosed by its characteristically diminished secretion and
Thrombin produces maximum ATP release through at least aggregation responses to all agonists except thrombin or a
two membrane binding sites. Collagen, ADP, and epineph- modest response to arachidonic acid.
rine test for abnormalities in their respective membrane
binding sites and the eicosanoid synthesis pathway. Arachi- Acquired Platelet Disorders. Platelets may become
donic acid tests for eicosanoid synthesis deficiencies. Ris either dysfunctional or hyperactive in acquired hematologic
tocetin checks for abnormalities of plasma VWF in VWD. and systemic disorders such as acute leukemias, aplastic
The following conditions may be detected through platelet anemias, myeloproliferative neoplasms, myelodysplastic syn-
lumiaggregometry. dromes, myeloma, uremia, liver disease, and chronic alcohol
CHAPTER 45 Laboratory Evaluation of Hemostasis 747

abuse. Investigation is made for these disorders in any case in measurement of these proteins is of diagnostic or prognostic
which aggregation is abnormal and no other explanation is significance is under investigation.50 Diagnostica Stago, Inc.
available. Platelet aggregometry may be used to predict the risk (Asnires, France), produces enzyme immunoassay kits for PF4
of bleeding or thrombosis in the acquired platelet functional and -thromboglobulin under the brand names Asserachrom
disorders.43 B-TG and Asserachrom PF4. Special collection techniques are
necessary, because PF4 and -thromboglobulin test results may
Testing for Heparin-Induced be invalidated by platelet activation during and even subse-
Thrombocytopenia with Thrombosis quent to specimen collection.51 CTAD tubes (see the section on
A description of the clinical manifestations and mechanism of hemostasis specimen collection) are required for specimen col-
heparin-induced thrombocytopenia with thrombosis (HIT) is lection for PF4 and -thromboglobulin assays. Plasma must
provided in Chapter 42 and a summary of aggregation-based undergo extraction before PF4 and -thromboglobulin assays
tests for HIT is provided in Chapter 44. Aggregation tests for are performed, because several eicosanoids cross-react with kit
HIT include light-transmittance aggregometry, washed platelet antibodies and falsely raise the results.52
light-transmittance aggregometry, washed platelet lumiag- Thromboxane A2, the active product of the eicosanoid
gregometry, whole-blood lumiaggregometry, and the washed pathway, has a half-life of 30 seconds, diffuses from the plate-
platelet carbon 14 (14C) serotonin release assay (SRA).44-46 All let, and spontaneously reduces to thromboxane B2, a stable,
these tests employ unfractionated heparin as their agonist. The measurable plasma metabolite (see Chapter 13). Efforts to
14
C SRA is available from specialized reference laboratories and produce a clinical assay for plasma thromboxane B2 have been
is regarded as the reference confirmatory method. Few local unsuccessful, because specimens must be collected in CTAD
institutions provide the 14C SRA, because a radionuclide license tubes to prevent in vitro platelet activation and must undergo
is required. Except for the 14C SRA, aggregometry and lumiag- an extraction step before the assay is performed. Thromboxane
gregometry tests for HIT have proven to be insensitive and have B2 is acted on by liver enzymes to produce an array of soluble
been largely discontinued. urine metabolites, including 11-dehydrothromboxane B2, which
is stable and measurable.53 Immunoassays of urine 11-
dehydrothromboxane B2 are used to characterize in vivo plate-
let activation.54 These assays require no special specimen
QUANTITATIVE MEASUREMENT
management and can be performed on random urine speci-
OF PLATELET MARKERS
mens. The urinary 11-dehydrothromboxane B2 assay also may
Immunoassay for the AntiPlatelet be used to monitor aspirin therapy and to identify cases of
Factor 4 (Heparin-Induced therapy failure or aspirin resistance.55
Thrombocytopenia) Antibody
Amiral etal developed an HIT screening immunoassay based
on their discovery that PF4 is the target for the heparin-
CLOT-BASED PLASMA
dependent antiplatelet antibody that causes HIT (see Chapter
PROCOAGULANT SCREENS
42).47 One adaptation of this principle is the GTI PF4 Enhanced
Solid Phase ELISA (GTI Diagnostics, Inc., Waukesha, Wisc.). The Lee-White whole-blood coagulation time test, described in
Patient plasma is incubated in microtiter plate wells that are 1913, was the first laboratory procedure designed to assess
coated with a solid-phase complex of purified PF4 and poly- coagulation.56 The Lee-White test is no longer used, but it was
sulfonate, a plastic molecule integral to plate construction that the first in vitro clot procedure based on the principle that the
resembles heparin. Heparin-dependent anti-PF4 antibodies time interval from the initiation of coagulation to visible clot
bind the PF4-polysulfonate complex. Bound antibodies are formation reflects the condition of the coagulation mecha-
detected using enzyme-conjugated antihuman immunoglo nism. A prolonged clotting time indicates coagulation inade-
bulin G (IgG), IgA, and IgM antibodies and a substrate chro- quacy. A 1953 modification, the activated clotting time (ACT)
mophore. This test is more sensitive than the 14C SRA and test, utilizes a particulate clot activator in the test tube, which
detects antibodies early in the development of HIT, but may speeds the clotting process. The ACT is still widely used in
detect antibodies that are unaccompanied by clinical symp- point-of-care instruments to monitor heparin therapy in high-
toms, known as biologic false positives.48 A second GTI kit that dosage applications, such as coronary artery bypass graft
uses only enzyme-conjugated antihuman IgG is more specific. surgery (see Chapters 46 and 47).
Competitors to the GTI kit employ PF4-heparin in place of The standard clinical profile of clot-based coagulation
PF4-polysulfonate and have similar sensitivity and specificity screening tests, which consists of the PT, PTT, fibrinogen assay,
characteristics. and thrombin clotting time (TCT), uses the clotting time prin-
ciple of the Lee-White test. Many specialized tests, such as
Assays for Platelet Activation Markers coagulation factor assays, tests of fibrinolysis, inhibitor assays,
Elevated plasma levels of the platelet-specific proteins - reptilase time, Russell viper venom time, dilute Russell viper
thromboglobulin and PF4 may accompany thrombotic stroke or venom time, and tests for circulating anticoagulants, also are
coronary thrombosis.49 The implication that in vivo platelet based on the relationship between time to clot formation and
activation contributes to the condition or that the coagulation function.
748 PART VIII Hemostasis and Thrombosis

Reagent

Membrane PL IX XI
Pre-K HMWK
Ca2+ XII
TF Thr XIIa

Ca2+ NCS

TF
VII VIIa XIa

Ca2+ PL

VIIIa VIII
IXa

Ca2+ PL Thr

Va V
X Xa

Ca2+ PL
XIII

Pro Thr

XIIIa

Fibrinogen Fibrin Cross-linked


fibrin
Figure 45-8 Prothrombin time (PT) reagent consists of tissue factor (TF), phospholipid (PL), and calcium. It activates the extrinsic and common pathways
of the coagulation mechanism beginning with factor VII. The PT is prolonged by deficiencies of factors VII, X, and V, prothrombin, and fibrinogen, when the
fibrinogen level is less than 100mg/dL. The PT is prolonged in warfarin therapy because warfarin suppresses production of factor VII, factor X, and prothrombin.
Factor VII has a short half-life (6 hours) and has the greatest effect on the PT. HMWK, High-molecular-weight kininogen (Fitzgerald factor); NCS, negatively
charged surface; Pre-K, prekallikrein (Fletcher factor); Pro, Prothrombin (factor II; zymogen); Thr, thrombin (activated factor II, or IIa; serine protease); Va, VIIIa,
activated factors V and VIII (serine protease cofactors); VIIa, IXa, Xa, XIa, activated factors VII, IX, X, XI, and (serine proteases); XIIa, activated factor XII (serine
protease, but not part of in vivo coagulation); XIIIa, activated factor XIII (transglutaminase).

Prothrombin Time moderately sensitive to factor V and X deficiencies, sensitive to


Prothrombin Time Principle severe fibrinogen and prothrombin deficiencies, and insensi-
PT reagents, often called thromboplastin or tissue thromboplastin, tive to deficiencies of factors VIII and IX.58,59 The PT is pro-
are prepared from recombinant or affinity-purified tissue factor longed in multiple factor deficiencies that include deficiencies
suspended in phospholipids mixed with a buffered 0.025- of factors VII and X and is used most often to monitor the
mol/L solution of calcium chloride.57 A few less responsive effects of therapy with the oral anticoagulant warfarin (see
thromboplastins are organic extracts of emulsified rabbit brain Chapter 46).
or lung suspended in calcium chloride. When mixed with
citrated PPP, the PT reagent triggers fibrin polymerization by Prothrombin Time Procedure
activating plasma factor VII (Figure 45-8). Calcium participates The tissue factorphospholipidcalcium chloride reagent is
in the formation of the tissue factorfactor VIIa complex, the warmed to 37 C. An aliquot of test PPP, 50 or 100mcL, is
factor VIIIafactor IXa complex, and the factor Vafactor Xa transferred to the reaction vessel, which also is maintained at
complex. The clot is detectable visually or by optical or elec- 37 C. The PPP aliquot is incubated at 37 C for at least 3 and
tromechanical sensors. Although the coagulation pathway for no more than 10 minutes. Aliquots that are incubated
implies that the PT would be prolonged in deficiencies of longer than 10 minutes deteriorate because of the rapid degra-
fibrinogen, prothrombin, and factors V, VII, VIII, IX, and X, dation of factor VIII. They are further affected by evaporation.
the procedure is most sensitive to factor VII deficiencies, A premeasured volume of reagent, 100 or 200mcL, is forcibly
CHAPTER 45 Laboratory Evaluation of Hemostasis 749
added to the PPP aliquot, and a timer is started. As the clot response to anticoagulant therapy has not stabilized. INRs are
forms, the timer stops, and the elapsed time is recorded. If the meant to be computed only for samples from patients with a
procedure is performed in duplicate, the duplicate values must stable anticoagulation response. During the first week of war-
be within 10% of their mean or the test is repeated for a third farin therapy, the physician should interpret PT results in
time. Most laboratory scientists perform PTs using automated seconds, comparing them with the reference interval. For a full
instruments that strictly control temperature, pipetting, and discussion of warfarin therapy monitoring, see Chapter 46.
interval timing. With automated instruments, duplicate testing
is unnecessary. Prothrombin Time Reference Interval
The PT reference interval varies from site to site depending on
Prothrombin Time Quality Control the patient population, type of thromboplastin used, type of
The medical laboratory scientist tests normal and prolonged instrument used, and pH and purity of the reagent diluent.
control PPP specimens at the beginning of each 8-hour shift Each center must establish its own range for each new lot of
or with each change of reagent. Although lyophilized control reagents or at least once a year. This may be done by testing a
PPPs are commercially available, the laboratory manager may sample of at least 20 specimens from healthy donors of both
choose to collect and pool PPP specimens from designated sexes spanning the adult age range over several days and com-
subjects to make home-brew controls. In this case, the speci- puting the 95% confidence interval of the results. A typical PT
mens must be collected and managed using the same tubes, reference interval is 12.6 to 14.6 seconds.
anticoagulant, and protocol that are used for patient plasma
specimen collection. The samples are pooled, tested, and ali- Use of Prothrombin Time as a Diagnostic Assay
quoted. Regardless of whether commercial or locally prepared The PT is performed diagnostically when any coagulopathy
controls are used, the control is tested alongside patient speci- is suspected. Acquired multiple deficiencies, such as disse
mens using the same protocol as for patient PPP testing. minated intravascular coagulation (DIC), liver disease, and
The normal control result should be within the reference vitamin K deficiency, all affect factor VII activity and are
interval and the prolonged control result should be within the detected through prolonged PT results. The PT is particularly
therapeutic range for warfarin. If the control results fall within sensitive to liver disease, which causes factor VII levels to
the stated limits in the laboratory protocol, the test results are become rapidly diminished (see Chapter 41).
considered valid. If the results fall outside the control limits, Vitamin K deficiency is seen in severe malnutrition, during
the reagents, control, and equipment are checked; the problem use of broad-spectrum antibiotics that destroy gut flora, with
is corrected; and the control and patient specimens are retested. parenteral nutrition, and in malabsorption syndromes. Vitamin
The operator records all the actions taken. Control results are K levels are low in newborns, in whom bacterial colonization
recorded and analyzed at regular intervals to determine the of the gut has not begun. Hemorrhage is likely in vitamin K
long-term validity of results. deficiency, and the PT is the best indicator. To distinguish
between vitamin K deficiency and liver disease, the laboratory
Reporting of Prothrombin Time Results and scientist determines both the factor V and factor VII level. Both
the International Normalized Ratio factor V and factor VII are reduced in liver disease; only factor
The medical laboratory scientist reports PT results to the nearest VII is reduced in vitamin K deficiency. Chapter 41 provides
tenth of a second along with the PT reference interval. If the details regarding liver disease and vitamin K deficiency.
PT assay is performed in duplicate, the results are averaged, and The PT is prolonged in congenital single-factor deficiencies
the average is reported. In view of the inherent variations of factor X, VII, or V; prothrombin deficiency; and fibrinogen
among thromboplastin reagents, most laboratories report the deficiency when the fibrinogen level is 100mg/dL or less.
international normalized ratio (INR) for patients with a stable When the PT is prolonged, but the PTT and TCT results are
anticoagulation response using the following formula60: normal, factor VII activity may be deficient. Any suspected
single-factor deficiency is confirmed with a factor assay. The PT
INR = (PTpatient PTgeometric mean of normal )ISI is not affected by factor VIII or IX deficiency, because the con-
centration of tissue factor in the reagent is high and those
where PTpatient is the PT of the patient in seconds, factors are bypassed in fibrin polymerization.
PTgeometric mean of normal is the PT of the geometric mean of the refer-
ence interval, and ISI is the international sensitivity index. Minimal Effectiveness of Prothrombin Time
Reagent producers generate the ISI for their thromboplastin by as a Screening Tool
performing an orthogonal regression analysis comparing its PT Preoperative PT screening of asymptomatic surgical patients to
results with the results of the international reference thrombo- predict intraoperative hemorrhage is not supported by preva-
plastin preparation (World Health Organization human brain lence studies, unless the patient is a member of a high-risk
thromboplastin). Most responsive thromboplastin reagents population.61,62 No clinical data support the use of the PT as a
have ISIs near 1, the assigned ISI of the World Health Organiza- general screening test for individuals at low risk, and the PT is
tion reagent. Automated coagulation timers request the reagent not useful for establishing baseline values in warfarin therapy.63
ISI from the operator and compute the INR for each test The therapeutic target range is based on the INR, not the base-
run, including runs analyzing specimens from patients whose line PT result or PT control value.
750 PART VIII Hemostasis and Thrombosis

TABLE 45-7 Factors That Interfere with the Validity of Partial Thromboplastin Time
Prothrombin Time (PT) Results Partial Thromboplastin Time Principle
The PTT (also called the activated partial thromboplastin time, or
Problem Solution
APTT) is performed to monitor the effects of unfractionated
Blood collection volume less PT falsely prolonged; recollect specimen. heparin therapy and to detect LA and specific anticoagulation
than specified minimum factor antibodies such as antifactor VIII antibody. The PTT is
Hematocrit 55% Adjust anticoagulant volume using also prolonged in all congenital and acquired procoagulant
formula and recollect. deficiencies except for deficiencies of factor VII or XIII.66
Clot in specimen All results unpredictably affected; The PTT reagent contains phospholipid (previously called
recollect specimen. partial thromboplastin) and a negatively charged particulate acti-
Visible hemolysis PT falsely shortened; recollect specimen. vator such as silica, kaolin, ellagic acid, or celite in suspension.
Icterus or lipemia Measure PT using a mechanical The phospholipid, which was historically extracted from rabbit
coagulometer. brain, is now produced synthetically. The activator provides a
Heparin therapy Use reagent known to be insensitive to surface that mediates a conformational change in plasma factor
heparin or one that includes a heparin XII that results in its activation (Figure 45-9). Factor XIIa forms
neutralizer such as Polybrene. a complex with two other plasma components, high-molecular-
Lupus anticoagulant PT result is invalid; use chromogenic weight kininogen (Fitzgerald factor) and prekallikrein (Fletcher
factor X assay instead of PT. factor). These three plasma glycoproteins, termed the contact
Incorrect calibration, incorrect Correct analytical error and repeat test. activation factors, initiate in vitro clot formation through the
dilution of reagents intrinsic pathway, but are not part of in vivo coagulation. Factor
XIIa, a serine protease, activates factor XI (XIa), which activates
factor IX (IXa) (see Chapter 40).
Factor IXa binds calcium, phospholipid, and factor VIIIa to
Limitations of Prothrombin Time form a complex. In the PTT reaction system, ionic calcium and
Specimen variations profoundly affect PT results (Table 45-7). phospholipid are supplied in the reagent. The factor IXa
The ratio of whole blood to anticoagulant is crucial, so col- calciumfactor VIIIaphospholipid complex catalyzes factor X
lection tubes must be filled to within tube manufacturers (Xa). Factor Xa forms another complex with calcium, phospho-
specifications and not underfilled or overfilled. Anticoagulant lipid, and factor Va, catalyzing the conversion of prothrombin
volume must be adjusted when the hematocrit is greater to thrombin. Thrombin catalyzes the polymerization of fibrin-
than 55% to avoid false prolongation of the results. Speci- ogen and the formation of the fibrin clot, which is the end
mens must be inverted five times immediately after collec- point of the PTT.
tion to ensure good anticoagulation, but the mixing must The factors whose deficiencies are associated with hemor-
be gentle. Clotted and visibly hemolyzed specimens are rhage and are reflected in prolonged PTT results, taken in the
rejected, because they give unreliable results. Plasma lipemia order of reaction, are XI, IX, VIII, X, and V; prothrombin; and
or icterus may affect the results obtained with optical fibrinogen, when fibrinogen is 100mg/dL or less. Most PTT
instrumentation. reagents are designed so that the PTT is prolonged when
Heparin may prolong the PT. If the patient is receiving the test PPP has less than approximately 0.3 units/mL (30%
therapeutic heparin, it should be noted on the order and com- of normal) of VIII, IX, or XI.67 The PTT also is prolonged
mented on when the results are reported. The laboratory in the presence of LA, an immunoglobulin with affinity for
manager selects thromboplastin reagents that are maximally phospholipid-bound proteins, and is prolonged by antifactor
sensitive to oral anticoagulant therapy and insensitive to VIII antibody, antibodies to factor IX and other coagulation
heparin. Many reagent manufacturers incorporate Polybrene factors, and therapeutic heparin. Factor VII and factor XIII defi-
into their thromboplastin reagent to neutralize heparin. The ciencies have no effect on the PTT. Deficiencies of factor XII,
medical laboratory scientist may detect unexpected heparin by prekallikrein, or high-molecular-weight kininogen prolong the
using the TCT test, which is described subsequently. PTT, but do not cause bleeding.
Some thromboplastins are prolonged by LAs. LAs are
members of the antiphospholipid antibody family and may Partial Thromboplastin Time Procedure
partially neutralize PT reagent phospholipids. Warfarin often To initiate contact activation, 50 or 100 mcL of warmed
is prescribed to prevent thrombosis in patients with LAs, but (37 C) reagent consisting of phospholipid and particulate
the PT may be an unreliable monitor of therapy in such cases. activator is mixed with an equal volume of warmed PPP. The
Patients who have an LA and are taking warfarin should be mixture is allowed to incubate for the exact manufacturer-
monitored using an alternative system, such as the chromo- specified time, usually 3 minutes. Next, 50 or 100mcL of
genic factor X assay.64,65 warmed 0.025mol/L calcium chloride is forcibly added to the
Reagents must be reconstituted with the correct diluents and mixture, and a timer is started. When a fibrin clot forms, the
volumes following manufacturer instructions. Reagents must timer stops, and the interval is recorded. Timing may be done
be stored and shipped according to manufacturer instructions with a stopwatch or by an automatic electromechanical or
and never used after the expiration date. photo-optical device. If the PTT is performed manually, the test
CHAPTER 45 Laboratory Evaluation of Hemostasis 751

Membrane PL IX XI
Pre-K HMWK
Ca2+ XII
TF Thr XIIa
Ca2+ NCS

TF
VII VIIa XIa

Ca2+ PL

VIIIa VIII Reagent


IXa

Ca2+ PL Thr

Va V
X Xa

Ca2+ PL
XIII

Pro Thr

XIIIa

Fibrinogen Fibrin Cross-linked


fibrin
Figure 45-9 Partial thromboplastin time (PTT) reagent consists of phospholipid (PL), a negatively charged particulate activator (NCS), and calcium. It
activates the intrinsic and common pathways of the coagulation mechanism through the contact factors XII, prekallikrein (Pre-K; also called Fletcher factor),
and high-molecular-weight kininogen (HMWK; also called Fitzgerald factor), none of which are significant in the in vivo coagulation mechanism. The PTT is
prolonged by deficiencies in Pre-K; HMWK; factors XII, XI, IX, VIII, X, and prothrombin; and fibrinogen, when the fibrinogen level is less than 100mg/dL. The
deficiencies for which the PTT reagent is specifically calibrated are factors VIII, IX, and XI. The PTT is prolonged in heparin therapy because heparin activates
plasma antithrombin, which neutralizes all the plasma serine proteases, particularly thrombin (IIa) and activated factor X (Xa). The PTT is prolonged in the
presence of lupus anticoagulant because the anticoagulant neutralizes essential reagent phospholipids. Pro, Prothrombin (factor II; zymogen); TF, tissue factor;
Thr, thrombin (activated factor II, or IIa; serine protease); Va, VIIIa, activated factors V and VIII (serine protease cofactors); VIIa, IXa, Xa, XIa, activated factors
VII, IX, X, XI, (serine proteases); XIIa, activated factor XII (serine protease, but not part of in vivo coagulation); XIIIa, activated factor XIII (transglutaminase).

should be done in duplicate, and the two results must match recorded and analyzed at regular intervals to determine the
within 10%. long-term validity of results.
Reagents must be reconstituted with the correct diluents and
Partial Thromboplastin Time Quality Control volumes following manufacturer instructions. Reagents must
The medical laboratory scientist tests normal and prolonged be stored and shipped according to manufacturer instructions
control plasma specimens at the beginning of each 8-hour shift and never used after the expiration date.
or with each new batch of reagent. More frequent use of con-
trols may be required by the laboratory director. Controls are Partial Thromboplastin Time Reference Interval
tested using the protocol for patient plasma testing. The PTT reference interval varies from site to site depending on
The normal control result should be within the reference the patient population, type of reagent, type of instrument, and
interval, and the abnormal control result should be within the pH and purity of the diluent. One medical center laboratory
therapeutic range for unfractionated heparin (see Chapter 46). has established 26 to 38 seconds as its reference interval. This
If the control results fall within the stated limits in the labora- range is typical, but each center must establish its own interval
tory protocol, the test results are considered valid. If the results for each new lot of reagent or at least once a year. This may
fall outside the control limits, the reagents, control, and equip- be done by testing a sample of 20 or more specimens from
ment are checked; the problem is corrected; and the control healthy donors of both sexes spanning the adult age range over
and patient specimens are retested. The operator records each several days and computing the 95% confidence interval of
control run and all the actions taken. Control results are the results.
752 PART VIII Hemostasis and Thrombosis

Monitoring of Heparin Therapy with Specific Factor Inhibitors


Partial Thromboplastin Time Specific inhibitors are IgG immunoglobulins directed against
Since the early 1970s, the PTT has been the standard method coagulation factors. Specific inhibitors arise in severe congeni-
for monitoring unfractionated heparin therapy, which is used tal factor deficiencies during factor concentrate treatment.
during coronary artery bypass graft surgery, during cardiac cath- Antifactor VIII, the most common of the specific inhibitors,
eterization, and in several medical conditions.68 The laboratory is detected in 10% to 20% of patients with severe hemophilia,
scientist establishes a PTT therapeutic range and publishes it to and antifactor IX is detected in 1% to 3% of factor IXdeficient
all inpatient units. A typical therapeutic range is 60 to 100 patients. Autoantibodies to factor VIII occasionally may be
seconds; however, the range varies widely and must be estab- present in individuals without hemophilia, usually in those
lished locally.69 The range must be reestablished with each with a postpartum bleeding syndrome or in older patients with
change of PTT reagent, including each lot change, and upon autoimmune disorders. Alloantibodies and autoantibodies to
instrument recalibration. Details on monitoring of heparin factor VIII are associated with severe anatomic hemorrhage.
therapy and establishment of the PTT therapeutic range are The presence of the latter is called acquired hemophilia (see
provided in Chapter 46. Chapter 41).

Use of Partial Thromboplastin Time Detection and Identification of Lupus Anticoagulants


as a Diagnostic Assay and Specific Inhibitors
The physician orders a PTT assay when a hemorrhagic disorder LA testing is part of every thrombophilia profile (see Chapter
is suspected or when recurrent thrombosis or the presence of 42) and is ordered by the physician when thrombophilia is
an autoimmune disorder points to the possibility of an LA.70 suspected. An unexpected prolonged screening PTT may also
The PTT result is prolonged when there is a deficiency of trigger an LA investigation. PTT mixing studies are necessary
one or more of the following coagulation factors: prothrom- for the initial detection of LAs.73 Mixing studies also distinguish
bin; factor V, VIII, IX, X, XI, or XII; or fibrinogen when LAs from specific inhibitors and factor deficiencies, and should
the fibrinogen level is 100 mg/dL or less. The PTT also is be available in all coagulation laboratories.74
prolonged in the presence of a specific inhibitor, such as When the initial PTT is prolonged beyond the upper limit
antifactor VIII or antifactor IX; a nonspecific inhibitor, of the reference interval, the laboratory scientist first deter-
such as LA; and interfering substances, such as fibrin degrada- mines if heparin is present by performing the TCT. A TCT result
tion products (FDPs) or paraproteins, which are present in that exceeds the upper limit of the TCT reference interval is
myeloma. evidence for the presence of heparin. In fact, heparin often
DIC prolongs PTT results because of consumption of pro- prolongs the TCT to 30 to 40 seconds. Heparin may be neutral-
coagulants, but the PTT results alone are not definitive for the ized using Polybrene or heparinase (Hepzyme; Siemens
diagnosis of DIC. Vitamin K deficiency results in diminished Healthcare Diagnostics), and the treated sample may be used
levels of procoagulant factors II, VII, IX, and X, and the PTT is for PTT mixing studies.
eventually prolonged. Because factor VII deficiency does not The heparin-free or heparin-neutralized patient plasma is
affect the PTT, however, and because it is the first coagulation then mixed 1:1 with reagent platelet-poor normal plasma (PNP;
factor to become deficient, the PTT is not as sensitive to vitamin see Figure 42-2). Several manufacturers make PNP, for example,
K deficiency or warfarin therapy as the PT. The PTT is not pro- frozen Cryocheck Normal Reference Plasma (Precision Bio-
longed in deficiencies of factor VII or XIII. No clinical data Logic, Inc., Dartmouth, Nova Scotia, Canada). A new PTT is
support the use of the PTT as a general screening test for indi- performed immediately on the 1:1 mixture. If the mixture
viduals at low risk.71 PTT corrects to within 10% of the PNP PTT (or to within the
reference interval) and the patient is experiencing bleeding, a
coagulation factor deficiency (coagulopathy) is presumed.
Partial Thromboplastin Time Mixing Studies Some LAs are time dependent and temperature dependent.
Lupus Anticoagulants Most antifactor VIII inhibitors are temperature-dependent
LAs are IgG immunoglobulins directed against several IgG4-class antibodies. If the immediate PTT corrects, a new
phospholipid-protein complexes.72 LAs prolong the phospholipid- mixture is prepared and incubated 1 to 2 hours at 37 C. If the
dependent PTT reaction. Most laboratories employ a moderate- incubated mixtures PTT fails to correct to within 10% of
phospholipid or high-phospholipid PTT reagent in their the incubated PNP PTT, presence of an inhibitor is likely. If
primary PTT assay to monitor heparin therapy and detect the patient is bleeding, a specific inhibitor such as antifactor
coagulopathies. Laboratories use a second low-phospholipid VIII is suspected, and a factor VIII activity assay is performed.
PTT reagent such as PTT-LA (Diagnostica Stago), more sensitive Although antifactor IX and other inhibitors have been docu-
to LA, as their LA screen (see Chapter 42). Because they have mented, antifactor VIII is the most common. The Bethesda
a variety of target antigens, LAs are called nonspecific inhibitors. titer procedure is used to confirm the presence of specific anti
Chronic presence of LAs confers a 30% risk of arterial or venous coagulation factor antibodies.
thrombosis; every acute care laboratory must provide a means If the PTT of the initial or incubated mixture fails to correct
for their detection. Together, chronic and transient LAs are and the patient is not bleeding, an LA is suspected, and an LA
found in 1% to 2% of unselected individuals. profile, as described in Chapter 42, is automatically ordered.
CHAPTER 45 Laboratory Evaluation of Hemostasis 753

Membrane PL IX XI
Pre-K HMWK
Ca2+ XII
TF Thr XIIa
Ca2+ NCS

TF
VII VIIa XIa

Ca2+ PL

VIIIa VIII
IXa

Ca2+ PL Thr

Va V
X Xa

Ca2+ PL
XIII

Pro Thr

Reagent XIIIa

Fibrinogen Fibrin Cross-linked


fibrin
Figure 45-10 Thrombin clotting time and reptilase time coagulation pathway. The reagent activates the coagulation pathway at the level of prothrombin
and tests for the polymerization of fibrinogen. HMWK, High-molecular-weight kininogen (Fitzgerald factor); NCS, negatively charged surface; Pre-K, prekallikrein
(Fletcher factor); PL, phospholipid; Pro, prothrombin (factor II; zymogen); TF, tissue factor; Thr, thrombin (activated factor II, or IIa; serine protease); Va, VIIIa,
activated factors V and VIII (serine protease cofactors); VIIa, IXa, Xa, XIa, activated factors VII, IX, X, XI (serine proteases); XIIa, activated factor XII (serine
protease, but not part of in vivo coagulation); XIIIa, activated factor XIII (transglutaminase).

LA profiles are available from tertiary care facilities and spe- and records the results. The normal control results should fall
cialty reference laboratories. within the laboratorys reference interval. The abnormal control
results should be prolonged to the range reached by the TCT
Thrombin Clotting Time in moderate hypofibrinogenemia. If the results fall outside the
Thrombin Clotting Time Reagent and Principle laboratory protocols control limits, the reagents, control, and
Commercially prepared bovine thrombin reagent at 2 National equipment are checked; the problem is corrected; and the
Institutes of Health (NIH) units/mL cleaves fibrinopeptides control is retested. The actions taken to correct out-of-limit
A and B from plasma fibrinogen to form a detectable fibrin tests are recorded. Control results are analyzed at regular inter-
polymer (Figure 45-10). vals (weekly is typical) to determine the longitudinal validity
of the procedure.
Thrombin Clotting Time Procedure
Reagent thrombin is warmed to 37 C for a minimum of 3 and Reporting of Thrombin Clotting Time Results
a maximum of 10 minutes. Thrombin deteriorates during incu- and Clinical Utility
bation and must be used within 10 minutes of the time incuba- A typical TCT reference interval is 15 to 20 seconds, although
tion is begun. An aliquot of normal PPP, usually 100mcL, is the reference interval should be established locally. The TCT is
also incubated at 37 C for a minimum of 3 minutes and a prolonged when the fibrinogen level is less than 100mg/dL
maximum of 10. The operator forcibly pipettes 200mcL of (hypofibrinogenemia) or in the presence of antithrombotic
thrombin into the PPP aliquot and starts a timer. TCT tests may materials such as FDPs, paraproteins, or heparin. Afibrinogen-
be performed in duplicate and the results averaged. emia (absence of fibrinogen) and dysfibrinogenemia (presence
of fibrinogen that is biochemically abnormal and nonfunc-
Thrombin Clotting Time Quality Control tional) also cause a prolonged TCT. Before a prolonged TCT
The medical laboratory scientist tests a normal control sample may be considered to be evidence of diminished or abnormal
and an abnormal control sample with each batch of TCT assays fibrinogen, the presence of antithrombotic substances, such
754 PART VIII Hemostasis and Thrombosis

as heparin, FDPs, or paraproteins, must be ruled out. The Procedure for the Fibrinogen Assay
TCT is part of the PTT mixing study protocol. It is used when- Thrombin Reagent. Most laboratory managers prefer
ever the PTT is prolonged to determine whether heparin is commercially manufactured diagnostic lyophilized bovine
present.75 thrombin reagent for fibrinogen assays. Pharmaceutical topical
The fibrinogen assay described later is a simple modification thrombin also may be used. The reagent is reconstituted accord-
of the TCT. In the fibrinogen assay, the concentration of reagent ing to manufacturer instructions and used immediately or ali-
thrombin is 50 NIH units/mL, or about 25 times that used in quoted and frozen. If thrombin is to be frozen, it should be
the TCT, and the patient specimen is diluted 1:10. This dilution prepared in a stock solution of 1000 NIH units/mL and frozen
minimizes the effects of heparin or antithrombotic proteins. at 70 C until it is ready for use. When thawed, thrombin is
The reptilase time procedure is identical to the TCT procedure stable for only a few hours and cannot be refrozen for later use.
except that the reptilase reagent is insensitive to the effects of
heparin. Calibration Curve. A calibration curve is prepared with
each change in reagent lots, reference plasma, or instrument.
Reptilase Time The laboratory scientist prepares the curve by reconstituting
Reptilase Time Reagent and Principle commercially available lyophilized fibrinogen calibration
Reptilase is a thrombinlike enzyme isolated from the venom plasma. Using Owren buffer, five dilutions of the calibration
of Bothrops atrox that catalyzes the conversion of fibrinogen to plasma are prepared: 1:5, 1:10, 1:15, 1:20, and 1:40. An
fibrin (Pefakit Reptilase Time; Pentapharm, Inc., Basel, Switzer- aliquot of each dilution, usually 200mcL, is transferred to each
land). In contrast to thrombin, this enzyme cleaves only fibri- of three reaction tubes or cups, warmed to 37 C, and tested
nopeptide A from the fibrinogen molecule, whereas thrombin by adding 100mcL of working thrombin reagent at 50 NIH
cleaves both fibrinopeptides A and B.76 The specimen require- units/mL. Time from addition of thrombin to clot formation
ments, procedure, and quality assurance protocol for the rep- is recorded, results of duplicate tests are averaged, and the
tilase time test are the same as those for the TCT. The reagent values in seconds are graphed against fibrinogen concentration
is reconstituted with distilled water and is stable for 1 month (Figure 45-11). Because patient PPPs are diluted 1:10 before
when stored at 1 C to 6 C. Reptilase time reagent is a poison testing, the 1:10 calibration plasma dilution is assigned the
that may be fatal if it directly enters the bloodstream. same fibrinogen concentration value as that of the undiluted
reconstituted calibration plasma.
Reptilase Time Clinical Utility
Reptilase is insensitive to heparin, but sensitive to dysfibrino- Test Protocol. The laboratory scientist prepares a 1:10
genemia, which profoundly prolongs the assay time. The rep- dilution of each patient PPP and control with Owren buffer.
tilase time test is also useful for detecting hypofibrinogenemia Then 200mcL of each of the diluted PPPs is warmed to 37 C
or dysfibrinogenemia in patients receiving heparin therapy. in each of two reaction tubes or cups for 3 minutes. After incu-
The reptilase time is prolonged in the presence of FDPs and bation, 100mcL of thrombin reagent is added, a timer is
paraproteins. started, and the mixture is observed until a clot forms. The
timer is stopped, values for duplicate runs are averaged, and
the interval in seconds is compared with the graph. Results are
COAGULATION FACTOR ASSAYS
reported in milligrams per deciliter of fibrinogen.
Fibrinogen Assay If the clotting time of the patient PPP dilution is short,
Principle of the Fibrinogen Assay indicating a fibrinogen level greater than 480mg/dL, a 1:20
The clot-based method of Clauss, a modification of the TCT, is dilution is prepared and tested. The resulting fibrinogen
the recommended procedure for estimating the functional
fibrinogen level.77,78 Reagent bovine thrombin is added to PPP, Seconds
catalyzing the conversion of fibrinogen to fibrin polymer. In 100
the fibrinogen assay, the thrombin reagent concentration is 50
NIH units/mL. The PPP to be tested is diluted 1:10 with Owren
60
buffer. Because of the concentrated reagent and diluted PPP, 120
there is an inverse relationship between the interval to clot 160
240
formation and the concentration of functional fibrinogen. The
10
relationship is linear when the fibrinogen concentration is
480
100 to 400mg/dL. Diluting the PPP also minimizes the anti
thrombotic effects of heparin, FDPs, and paraproteins: heparin
levels less than 0.6 units/mL and FDP levels less than
100mcg/dL do not affect the results of the fibrinogen assay
provided the fibrinogen concentration is 150mg/dL or greater. 1
The interval to clot formation is compared with the results 10 100 1000
for reference plasma dilutions. A reference curve is prepared in Fibrinogen concentration in mg/dL
each laboratory and updated regularly. Figure 45-11 Fibrinogen calibrator curve plotted on log-log axes.
CHAPTER 45 Laboratory Evaluation of Hemostasis 755

concentration from the graph must be multiplied by 2 to com- therapy, LA, or a factor-specific inhibitor, the medical laboratory
pensate for the dilution. If the clotting time of the original 1:10 scientist may suspect a congenital single-factor deficiency. Three
patient PPP dilution is prolonged, indicating less than 200mg/ factor deficiencies that give this reaction pattern and cause hem-
dL of fibrinogen, a 1:5 dilution is prepared. The resulting con- orrhage are factor VIII deficiency (hemophilia A), factor IX
centration reading from the graph is divided by 2 to compen- deficiency (hemophilia B), and factor XI deficiency, which
sate for the greater concentration of the specimen. causes a mild intermittent bleeding disorder called Rosenthal
syndrome found primarily in Ashkenazi Jews.80 These deficiencies
Fibrinogen Assay Quality Control are most often detected in childhood. The next step in diagnosis
All results for duplicate tests must agree within a coefficient of of a congenital single-factor deficiency is the performance of a
variation of less than 7%. The medical laboratory scientist tests a one-stage single-factor assay based on the PTT system.
normal control sample and an abnormal control sample with Although necessary for diagnosis, PTT-based single-factor
each batch of specimens for which fibrinogen levels are measured assays are most often performed on specimens from patients
and records the results. The normal control results should be with previously identified single-factor deficiencies. Their
within the laboratorys reference interval. The abnormal control purpose is to monitor supportive therapy during bleeding epi-
results should be less than 100mg/dL. If either control result falls sodes or invasive procedures. Because hemophilia A is the most
outside the control limits, the reagents, control, and equipment common single-factor deficiency disorder, this discussion is
are checked; the problem is corrected; and the control is retested. confined to the factor VIII assay; however, the protocol may be
The actions taken to correct out-of-limit tests are recorded. generalized to the assays for factors IX and XI.
Control results are analyzed at regular intervals (weekly is The medical laboratory scientist uses the PTT system to esti-
typical) to determine the longitudinal validity of the procedure. mate the concentration of functional factor VIII by incorporat-
ing commercially prepared factor VIIIdepleted PPP in the
Results and Clinical Utility of the Fibrinogen Assay test system (Cryocheck Factor VIII Deficient Plasma; Precision
One institutions reference interval for fibrinogen concentration BioLogic). Most factor VIIIdepleted plasma is PPP collected
is 220 to 498mg/dL, although each local institution prepares from normal donors and immunodepleted using monoclonal
its own interval. Hypofibrinogenemia, a fibrinogen level of less antifactor VIII antibody bound to a separatory column.81
than 220mg/dL, is associated with DIC and severe liver disease. In the PTT system, factor VIIIdepleted PPP provides normal
Moderately severe liver disease, pregnancy, and a chronic activity of all procoagulants but factor VIII. Tested alone,
inflammatory condition may cause an elevated fibrinogen level, factor VIIIdepleted PPP has a prolonged PTT, but when
greater than 498mg/dL. Congenital afibrinogenemia leads to normal PPP is added, the PTT reverts to normal. In contrast, a
prolonged clotting times and is associated with a variable hem- prolonged result for a mixture of patient PPP and factor VIII
orrhagic disorder. Dysfibrinogenemia may give the same results depleted PPP implies that the patient PPP is factor VIII defi-
as hypofibrinogenemia by this test method, because some cient. The clotting time interval for the mixture of patient PPP
abnormal fibrinogen species are hydrolyzed more slowly by and factor VIIIdepleted PPP may be compared with a previ-
thrombin than is normal fibrinogen. Some forms of dysfibrino- ously prepared reference curve to estimate the level of factor
genemia may be associated with thrombosis.79 VIII activity in the patient PPP. The quantitative factor assay is
Fibrinogen values measured using immunologic assays and typically performed on three or four dilutions of patient PPP
turbidimetric methods (Ellis-Stransky technique; PT-Fibrinogen for instance, 1:10, 1:20, 1:40, and 1:80and the results com-
HS Plus, Beckman Coulter, Inc., Brea, Calif.) are normal in pared with a reference curve.
dysfibrinogenemia. The fibrinogen concentration is estimated
from reaction mixture turbidity and reported with each PT. Factor VIII Assay Reference Curve
To prepare a reference curve for the factor VIII assay, the labora-
Limitations of the Fibrinogen Assay tory scientist obtains a reference plasma such as CAP FVIIIc RM
Although antithrombotic effects are minimized by the dilution (College of American Pathologists, Northfield, Ill.) and pre-
of PPP specimens, heparin levels greater than 0.6 units/mL and pares a series of dilutions with buffered saline.82 Although
FDP levels greater than 100mcg/mL prolong the results and laboratory protocols vary, most laboratory scientists prepare a
give falsely lowered fibrinogen results. Care must be taken that series of five dilutions, from 1:5 to 1:500. Each dilution is
the thrombin reagent is pure and has not degenerated. Expo- mixed with reagent factor VIIIdepleted plasma and tested in
sure to sunlight or oxidation results in rapid breakdown. The duplicate using the PTT system. The duplicate results are aver-
working dilution lasts only 1 hour at 1 C to 6 C and should aged and plotted on log-log or log-linear graph paper (Figure
remain cold until just before testing. 45-12). The 1:10 dilution is assigned the factor VIII assay activ-
ity value found on the package insert. When patient PPP is
Single-Factor Assays Using the Partial tested, the time interval obtained is entered on the vertical
Thromboplastin Time Test coordinate and converted to a percentage.
Principle of Single-Factor Assays Based on
Partial Thromboplastin Time Factor VIII Assay Procedure
If the PTT is prolonged and the PT and TCT are normal, and there The medical laboratory scientist (or the automated coagulom-
is no ready explanation for the prolonged PTT such as heparin eter) prepares 1:10, 1:20, 1:40, and 1:80 dilutions of each
756 PART VIII Hemostasis and Thrombosis

Seconds with each assay and records the results. The normal control
100 results should fall within the reference interval. The deficient
1.7 control results should be in the range of 10% factor VIII activity
90
or below. If either control result falls outside the control limits,
80
the reagents, control, and equipment are checked; the problem
70 is corrected; and the control is retested. The scientist records
17
all actions taken to correct out-of-limit tests. Control results are
60
43 analyzed at regular intervals (weekly is typical) to determine
50 86 the longitudinal validity of the procedure.
40 172
Limitations of Single-Factor Assays
30 Interlaboratory coefficients of variation for the factor VIII assay
1 10 100 1000
reach 80%, which implies undesirable variation in the inter-
% Activity
pretation of therapeutic monitoring results from unrelated
Figure 45-12 Factor VIII assay calibrator curve plotted on linear-log axes.
institutions. To reduce inherent variation, the medical labora-
tory scientist uses an assayed commercial plasma to prepare the
patient PPP and control specimen, then mixes each dilution reference curve and selects reference dilutions that correspond
with equal volumes of factor VIIIdepleted plasma and PTT to only the linear portion of the curve. The laboratory must
reagent. In most cases, 100mcL of PTT reagent is mixed with assay three or more dilutions of patient PPP to check for inhibi-
100mcL each of patient PPP dilution and factor VIIIdepleted tors. The scientist also selects a matching reagent-instrument
plasma mixture. All dilutions of each specimen or control system with a demonstrated coefficient of variation of less than
are tested in duplicate. After incubation at 37 C for the 5% and uses factor-depleted substrates with no trace of the
manufacturer-specified time, typically 3 minutes, 100mcL of depleted factor.83 As with the PTT test, good specimen manage-
0.025mol/L calcium chloride is added, and a timer is started. ment is essential. Clotted, hemolyzed, icteric, or lipemic speci-
The interval is recorded in seconds, duplicates are averaged, the mens are rejected, because they give unreliable results. Reagents
mean result is compared with the reference curve, and the must be reconstituted with the correct diluents and volumes
percentage of factor VIII activity is reported. Factor activity following manufacturer instructions. Reagents must be stored
results for the 1:20, 1:40, and 1:80 dilutions are multiplied and shipped in accordance with manufacturer instructions and
by 2, 4, and 8, respectively, to compensate for the dilutions and never used after the expiration date.
should match the results of the 1:10 dilutions within 10%. If
the results of the dilutions do not match within 10%, they are Bethesda Titer for AntiFactor VIII Inhibitor
considered to be nonparallel. An LA may be present, and the The Bethesda titer is used to confirm the presence of and quan-
assay cannot provide a reliable estimate of factor VIII activity. tify an antifactor VIII inhibitor, which is typically an IgG4-class
Tests for factors IX and XI are performed using the same immunoglobulin.84 In this method, 200mcL of patient PPP is
approach except that the appropriate factor-depleted plasma is incubated with 200mcL of reagent PNP for 2 hours at 37 C.
substituted for factor VIIIdepleted plasma. Tests for the A control specimen consisting of 200mcL of imidazole buffer
contact factors XII, prekallikrein, and high-molecular-weight at pH 7.4 mixed with 200mcL of reagent normal PPP is incu-
kininogen are seldom requested, because deficiencies are not bated simultaneously. During the incubation period, anti
associated with bleeding disorders. Acquired and congenital factor VIII from the patient PPP neutralizes a percentage of PNP
contact factor deficiencies are relatively common, however, and factor VIII activity. The degree of factor VIII activity neutralized
cause PTT prolongation. Factor XII, prekallikrein, and high- is proportional to the level of inhibitor activity. After incuba-
molecular-weight kininogen assays are available from hemo- tion, residual factor VIII activity in the patient PPPPNP mixture
stasis reference laboratories, and their use may be necessary to is measured using specific factor activity assay as described in
account for an unexplained prolonged PTT. the section on factor assays using the PTT system.
The titer of inhibitor is expressed as a percentage of the
Expected Results and Clinical Utility control. If the patient PPPPNP mixture retains 75% of the
of Single-Factor Assays residual factor VIII activity of the control, no factor VIII inhibitor
The reference interval for factor VIII activity is 50% to 186%. is present. If the residual factor VIII level is 25% that of the
Spontaneous symptoms of hemophilia are evident at activity control, the patient PPP factor VIII inhibitor level is titered using
levels of 10% or less. The test is used most often to estimate several dilutions of the patient specimen in reagent normal PPP.
the plasma level of factor VIII activity during therapy (see One Bethesda unit of activity is the amount of antibody that
Chapter 41). Chronically elevated factor VIII predicts an ele- leaves 50% residual factor VIII activity in the mixture.
vated risk of venous thrombotic disease (see Chapter 42).
Single-Factor Assays Using
Single-Factor Assay Quality Control the Prothrombin Time Test
All duplicate results must agree within 10%. The medical labo- If the PTT and the PT are both prolonged, the TCT is normal,
ratory scientist tests a normal and a deficient control specimen and there is no ready explanation for the prolonged test results,
CHAPTER 45 Laboratory Evaluation of Hemostasis 757

TABLE 45-8 Factor Assays Using the TCT, PT, and PTT in infants, with seepage at the umbilical stump.87 In adults,
Test Systems bleeding is slow but progressive, accompanied by poor wound
healing and slowly resolving hematomas. Recurrent spontane-
Factor System
ous abortion and posttraumatic hemorrhage are common.
Fibrinogen (I) Modified thrombin clotting time Acquired factor XIII inhibitors cause severe bleeding that does
Prothrombin (II) Prothrombin time not respond to therapy.
Factor V Prothrombin time
Factor VII Prothrombin time Principle and Procedure of the Factor XIII
Factor VIII Partial thromboplastin time Qualitative Test (5-mol/L Urea Solubility)
Factor IX Partial thromboplastin time When a patient comes for treatment of bleeding and poor
Factor X Prothrombin time wound healing and the PTT, PT, platelet count, and fibrinogen
Factor XI Partial thromboplastin time level are normal, the scientist may consider the time-honored
Factor XIII Chromogenic assay qualitative 5-mol/L urea solubility test for factor XIII. The
unstable clot that forms in factor XIII deficiency or in the pres-
PT, Prothrombin time; PTT, partial thromboplastin time; TCT, thrombin clotting time. ence of factor XIII inhibitor dissolves in a 5-mol/L urea solu-
tion, whereas a factor XIIIastabilized clot remains intact for
at least 24 hours.
such as liver disease, vitamin K deficiency, DIC, or oral antico- The medical laboratory scientist prepares three tubes. Tube
agulant (warfarin) therapy, the medical laboratory scientist 1 receives 300mcL of patient PPP, and tube 3 receives 300mcL
may suspect a congenital single-factor deficiency. Three rela- of PNP. Tube 2 receives 200mcL of patient PPP and 100mcL
tively rare factor deficiencies that give this reaction pattern and of normal PPP to test for the presence of an inhibitor. The
cause hemorrhage are prothrombin deficiency, factor V defi- scientist pipettes 100mcL of 0.025mol/L calcium chloride
ciency, and factor X deficiency. If the PT is prolonged and all into each tube. After clot formation, all three tubes are incu-
other test results are normal, factor VII deficiency is suspected. bated at 37 C for 30 minutes. Next, 3mL of 5-mol/L urea
The next step is performance of a one-stage single-factor assay solution is transferred to each tube, and the tubes are tapped
based on the PT test system, which is a relatively rare event. gently to dislodge the clots from the sides. The tubes are
The principles and procedure described in the section on capped and incubated at room temperature for 24 hours, but
single-factor assay using the PTT system may be applied except are observed for evidence of clot disintegration at 1, 2, 4,
that PT reagent replaces the PTT reagent in the test system, and and 24 hours. Decreasing size of clot, fragmentation, and
the PT protocol is followed. Factor II (prothrombin)depleted, increasing turbidity of the urea solution are evidence of clot
factor Vdepleted, factor VIIdepleted, and factor Xdepleted disintegration. The clot in the patient plasma tube (tube 1) is
plasmas are available (Table 45-8). compared with the clot in the normal plasma tube (tube 3),
and results are reported as factor XIII present or factor XIII
Factor XIII Assay: 5-mol/L Urea Solubility absent. If a factor XIII inhibitor is present in the patient
Coagulation factor XIII is a transglutaminase that catalyzes plasma, the clot dissolves in tube 1, the patient tube, and in
covalent cross-links between the and chains of fibrin the second tube. A clot in normal PPP remains intact for
polymer.85 Cross-linking strengthens the fibrin clot and renders 24 hours.
it resistant to proteases. This is the final event in plasma coagu-
lation, and it is essential for normal hemostasis and normal Quantitative Factor XIII Assays
wound healing. Plasma, platelet, and tissue factor XIII function Several manufacturers provide quantitative factor XIII activity
identically. Neither the PT nor the PTT is prolonged by factor assays that are rapidly replacing the 5-mol/L urea assay. In one
XIII deficiency. such assay, the Technochrom Factor XIII (DiaPharma Group,
Inherited factor XIII deficiency, an autosomal recessive dis- Inc., West Chester, Ohio), quantitation of factor XIII activity is
order, affects both sexes in all races. The first report of the based on the measurement of ammonia released during the in
deficiency appeared in 1960, and the frequency is estimated at vitro transglutaminase reaction. Plasma factor XIII is activated
1 in 2 million. Factor XIII levels also may be low in chronic by reagent thrombin. The resultant factor XIIIa then cross-
DIC secondary to Crohn disease, leukemias, ulcerative colitis, links the fibrin amine substrate glycine ethyl ester to the
sepsis, inflammatory bowel disease, surgery, and Henoch- glutamine residue of a peptide substrate, releasing ammonia.
Schnlein purpura. In these cases, the factor XIII level decreases The concentration of ammonia is monitored in a glutamate
to 50% of normal, not low enough to create symptoms, dehydrogenasecatalyzed reaction dependent on NADPH (the
although occasionally acquired factor XIII deficiencies produce reduced form of nicotinamide adenine dinucleotide phos-
low enough levels to cause mild bleeding. Acquired factor XIII phate). Consumption of NADPH is measured by the decrease
inhibitors have been described in patients treated with isonia- of absorbance at 340nm. The absorbance is inversely pro
zid, penicillin, valproate, and phenytoin.86 These drugs may portional to factor XIII activity. Several manufacturers market
cause complete absence of factor XIII. immunoassays for factor XIII, which provide factor XIII
Factor XIII activity levels lower than 5% result in hemor- concentration but do not identify functional factor XIII
rhage. In congenital factor XIII deficiency, bleeding is evident abnormalities.
758 PART VIII Hemostasis and Thrombosis

Qualitative D-Dimer Assay


TESTS OF FIBRINOLYSIS
The automated quantitative D-dimer assay has largely replaced
Quantitative D-Dimer Immunoassay manual D-dimer or FDP assays. The SimpliRED D-dimer assay
Physiology of Fibrin Degradation Products (BBInternational, Inc., Dundee, United Kingdom) is a manual
and D-Dimers method that uses visible latex particles coated with mono
During coagulation, fibrin polymers become cross-linked by clonal antibody. The SimpliRED D-dimer assay is suited to
factor XIIIa and simultaneously bind plasma plasminogen and low-volume or near-patient (point-of-care) applications. The
tissue plasminogen activator (TPA) (see Chapter 40). Over manufacturer reports the clinical sensitivity for pulmonary
several hours, bound TPA activates nearby plasminogen to form embolus to be 94% and the specificity to be 67%.95
plasmin. The bound plasmin cleaves fibrin and yields the FDPs
D, E, X, and Y and D-dimer. The FDPs represent original fibrino- Fibrin Degradation Product Immunoassay
gen domains, and D-dimers are covalently linked D domains Although the FDP assay has largely been replaced by the auto-
reflecting the cross-linking effects of factor XIIIa. Assays for mated quantitative D-dimer assay or the manual semiquantita-
FDPs, including D-dimer, are convenient for detecting active tive D-dimer assay, FDPs may be detected using a semiquantitative
fibrinolysis, which indirectly implies the occurrence of throm- visible agglutination immunoassay.96 One such method is the
bosis. Normally, FDPs, including D-dimer, circulate at concen- 1972 Thrombo-Wellcotest (Remel, Inc., Lenexa, Kan.).97 Poly-
trations of less than 2ng/mL. Fibrinolysis yields FDPs and styrene latex particles in buffered saline are coated with poly-
D-dimer at concentrations greater than 200ng/mL. Increased clonal antibodies specific for D and E fragments calibrated to
FDP and D-dimer concentrations are characteristic of acute and detect FDPs at a concentration of 2mcg/mL or greater. The
chronic DIC, systemic fibrinolysis, deep vein thrombosis, and assay usually is performed on serum collected in special tubes
pulmonary embolism.88 FDPs, including D-dimer, also are that promote clotting and prevent in vitro fibrinolysis, although
detected in plasma after thrombolytic therapy.89 plasma-based assays are also available.

Principle of the Quantitative D-Dimer Assay Plasminogen Chromogenic Substrate Assay


Plasma D-dimer immunoassays abound, and several diagnos- Plasminogen, the precursor of the trypsinlike proteolytic enzyme
tics distributors offer automated quantitative immunoassays plasmin, is produced in the liver and circulates as a single-chain
for plasma D-dimers that generate results within 30 minutes.90 glycoprotein (see Chapter 40). When bound to fibrin, plasmino-
Microlatex particles in buffered saline are coated with mono- gen is converted to plasmin by the action of nearby TPA. Bound
clonal antiD-dimer antibodies. The coated particles are agglu- plasmin degrades fibrin, whereas free plasmin is rapidly inacti-
tinated by patient plasma D-dimer; the resultant turbidity is vated by a circulating inhibitor, 2-antiplasmin.
measured using turbidometric or nephelometric technology. Congenital plasminogen deficiencies are associated with
Sensitivity varies depending on the avidity of the monoclonal thrombosis in some families.98 Acquired plasminogen deficien-
antiD-dimer and the detection method; however, most cies are seen in DIC and acute promyelocytic leukemia.99
methods detect concentrations as low as 10ng/mL. Thrombolytic therapy is ineffective when plasminogen activity
is low. Plasminogen is readily measured in PPP using a chro
Clinical Value of the Quantitative D-Dimer Assay mogenic substrate assay, available from several manufacturers.
The quantitative D-dimer assay is essential for ruling out
venous thromboembolic disease in patients with low pretest Principle of the Plasminogen Chromogenic
probability and is required for detecting and monitoring Substrate Assay
DIC (see Chapter 42).91 The D-dimer assay helps rule out Chromogenic substrates employ synthetic polypeptides whose
acute myocardial infarction and ischemic stroke and may be amino acid sequences are designed to be specific for particular
used to monitor the efficacy of long-term warfarin therapy.92 enzymes.100 Plasmin hydrolyzes a bond in the polypeptide
The various quantitative D-dimer assays have negative predic- sequence valine-leucine-lysine (Val-Leu-Lys). A fluorophore or
tive values of 90% to 95% and may be used to rule out deep a chromophore such as para-nitroaniline (pNA) is covalently
vein thrombosis and pulmonary thrombotic emboli in patients bound to the carboxyl end of the polypeptide and may be
at low risk without resorting to compression ultrasonography, released on digestion. S-2251, composed of H-D-Val-Leu-
tomography, or venous imaging.93,94 Because of the high sen- Lys-pNA, is a chromogenic substrate for plasmin. On plasmin
sitivity but low specificity (60% to 70%) of the quantitative digestion, the pNA is released and turns yellow (Figure 45-13).
D-dimer assay, physicians do not use this assay to positively Streptokinase is an exogenous plasminogen activator derived
diagnose venous thromboembolic disease, but only to rule it from cultures of -hemolytic streptococci. Streptokinase is
out. Because any chronic or acute inflammation is accompa- added to patient PPP, where it binds and activates plasminogen.
nied by elevated D-dimer concentrations, the assay cannot be The resulting streptokinase-plasmin complex reacts with a chro-
used to rule in thromboembolic disease. The upper limit mogenic substrate such as S-2251 to release a color whose
of the reference interval for the quantitative D-dimer assay intensity is proportional to the plasminogen concentration.
varies with the methodology, ranging from 250ng/mL to Several analogous chromogenic and fluorogenic substrates are
500ng/mL. In DIC, D-dimer levels may reach 10,000 to suitable for plasminogen measurement. A control plasma is
20,000ng/mL. tested with the patient plasma, and the results are recorded.
CHAPTER 45 Laboratory Evaluation of Hemostasis 759

Plasminogen + Streptokinase Plasmin:Streptokinase TPA


Plasminogen Plasmin
Soluble fibrin
Plasmin:Streptokinase
R-pNA R-COOH + pNA (color) Plasmin
R-pNA R-COOH + pNA (color)
Figure 45-13 Assay of plasma plasminogen using the chromogenic sub-
strate method. Reagent streptokinase activates plasminogen to form plasmin. Figure 45-14 To assay tissue plasminogen activator (TPA), plasma con-
R-pNA designates a chromogenic substrate, where R indicates several taining TPA is added to plasminogen to produce plasmin. Plasmin activity is
choices of peptide sequence and pNA (para-nitroaniline) is the chromophore. measured using the same chromogenic substrate system as in the plasmino-
In the case of plasminogen, the R represents the peptide sequence valine- gen assay. The intensity of color is proportional to TPA activity. pNA, Para-
leucine-lysine-pNA (Val-Leu-Lys-pNA). The Val-Leu-Lys amide sequence is nitroaniline; R, variable peptide sequence.
recognized and hydrolyzed by plasmin.
rest, tourniquet application should be minimal, and immedi-
ate acidification of the specimen in acetate buffer is neces-
Results and Clinical Utility of the Plasminogen sary.107 Acidification may be accomplished using the Stabilyte
Chromogenic Substrate Assay acidified citrate tube (Tcoag US, Inc, Berkeley Heights, NJ).
The plasminogen reference interval is 5 to 13.5mg/dL. Plas- Supernatant PPP may be frozen at 70 C until the assay is
minogen levels are decreased in thrombolytic therapy, DIC, performed.
hepatitis, and cancer. Hereditary deficiencies also have been
recorded.101 Decreased plasminogen is associated with throm- Principle of the Tissue Plasminogen Activator Assay
bosis. Plasminogen rises in inflammation and during preg- The level of TPA antigen may be estimated by enzyme immuno
nancy, and high levels may be associated with hemorrhage. assay. To measure TPA activity, a specified concentration of
Plasminogen levels are elevated in systemic fibrinolysis.102 Plas- reagent plasminogen is added to the patient plasma (Chro-
minogen assays are seldom offered in acute care facilities but molyse TPA Activity; Tcoag US). Plasma TPA activates the plas-
are readily available at specialty reference laboratories. minogen, and the resultant plasmin activity is measured using
a chromogenic substrate. The resulting color intensity is pro-
Tissue Plasminogen Activator Assay portional to TPA activity (Figure 45-14). The system may incor-
Physiology of Tissue Plasminogen Activator porate soluble fibrin to increase TPA activity.
The two physiologic human plasminogen activators are TPA
and urokinase. TPA is synthesized in vascular endothelial cells Plasminogen Activator Inhibitor 1 Assay
and released into the circulation, where its half-life is approxi- PAI-1 is produced by vascular endothelial cells and hepatocytes
mately 3 minutes and its plasma concentration averages 5ng/ and circulates in plasma bound to vitronectin at an average
mL.103 Urokinase is produced in the kidney and vascular endo- concentration of 10mcg/L with diurnal variations.108 An inac-
thelial cells and has a half-life of approximately 7 minutes and tive form of PAI-1 circulates in high concentrations in plate-
a concentration of 2 to 4ng/mL.104 Both activators are serine lets.109 PAI-1 inactivates free TPA by covalent binding. Elevated
proteases that form ternary complexes with bound plasmino- PAI-1 is associated with venous thrombosis and may be a
gen at the surface of fibrin, activating the plasminogen and cardiovascular risk factor (see Chapter 42). A few cases of
initiating thrombus degradation. Both are covalently inacti- PAI-1 deficiency have been reported; however, hemorrhage
vated by the endothelial secretion plasminogen activator inhibitor apparently occurs only in the complete absence of PAI-1.
1 (PAI-1). Blood is collected from patients at rest into an acidified
citrate tube (Stabilyte; Tcoag US) and centrifuged immediately
Clinical Significance of Tissue Plasminogen Activator to make PPP; this avoids in vitro release of platelet PAI-1.
The reference interval upper limit for TPA activity is 1.1 units/ Several immunometric and chromogenic substrate methods
mL, and the upper limit for TPA antigen is 14ng/mL. TPA is are available for estimation of PAI-1 antigen and PAI-1 activity.
the primary mediator of fibrinolysis and is the model for syn- One enzyme immunoassay for functional PAI-1 uses urokinase
thetic TPA (Activase; Genentech, Inc., South San Francisco, to bind PAI-1. The urokinasePAI-1 complex is immobilized
Calif.). Decreased TPA levels may indicate increased risk with solid-phase monoclonal antiPAI-1 and is measured
of myocardial infarction, stroke, or deep vein thrombosis, using monoclonal antiurokinase as the detecting antibody.110
although more data are needed to verify a relationship.105 Most chromogenic substrate kits for PAI-1 use an indirect
Impaired fibrinolysis in the form of TPA deficiency or PAI-1 measurement approach (Spectrolyse PAI-1; Tcoag US). Patient
excess also is associated with deep vein thrombosis and myo- PPP is mixed with a measured amount of reagent TPA. Residual
cardial infarction.106 TPA is assayed in the plasminogen system as shown in Figure
45-15. The resulting color intensity is inversely proportional to
Specimen Collection for the Tissue Plasminogen plasma PAI-1 activity.
Activator Assay Confirmation of complete PAI-1 deficiency may be accom-
TPA activity exhibits diurnal variation and rises upon exercise. plished using the serum PAI-1 assay. In serum, platelet PAI-1 is
Further, TPA is unstable in vitro because it rapidly binds PAI-1 expressed in excess. In true PAI-1 absence, no PAI-1 is detect-
after collection. For specimen collection, patients should be at able in serum.
760 PART VIII Hemostasis and Thrombosis

to 10 hours. The time required for the intrinsic plasmin to lyse


TPA + PAI-1 TPA: PAI-1 + Residual TPA
the fibrin clot in hours is the ELT.

Residual TPA
Plasminogen Plasmin Results of Euglobulin Clot Lysis Time Test
Soluble fibrin
If the euglobulin clot dissolves in less than 2 hours, excess
Plasmin fibrinolysis is present. If the clot remains for longer than 10
R-pNA R-COOH + pNA (color) hours, the fibrinolytic pathway is deficient.
Figure 45-15 To assay plasminogen activator inhibitor 1 (PAI-1) activity,
plasma containing PAI-1 is added to reagent tissue plasminogen activator Limitations of Euglobulin Clot Lysis Time Test
(TPA) of known concentration. The residual TPA is assayed as shown in Positive and negative plasma control specimens are included.
Figure 45-14. The intensity of color is inversely proportional to PAI-1 level. PNP is used as the negative control, whereas PNP plus strepto-
pNA, Para-nitroaniline; R, variable peptide sequence. kinase (a plasminogen activator) is used as the positive control.
The rate at which the clot disappears in the patient fraction is
compared with the rate of clot disappearance in the positive
and negative control specimens.
Euglobulin Clot Lysis Time Test The scientist prepares another control, the patient-
Excessive fibrinolytic activity occurs in a variety of conditions. activated control. This is an aliquot of the patients euglobulin
Inflammation and trauma may be reflected in a radical increase fraction with streptokinase added. This control ensures against
in circulating plasmin that has the potential to cause hemor- invalid results caused by depletion of patient plasminogen.
rhage. Bone trauma, fractures, and surgical dissection of bone, When patient plasminogen levels are diminished, such as
as in cardiac surgery, may raise fibrinolysis activity.111 Fibrino- in long-term DIC, clot dissolution extends beyond 10 hours.
lysis deficiencies occur when TPA or plasminogen levels become The patient-activated control indicates when this condition
depleted, or when excess secretion of PAI-1 depresses TPA activ- exists, because streptokinase-activated patient plasmin causes
ity. A time-honored approach to measurement of fibrinolytic rapid clot dissolution. When plasminogen is depleted, the
activity is the euglobulin clot lysis time (ELT) test. streptokinase-activated control produces no dissolution, and
the ELT is prolonged.
Principle and Procedure of the Euglobulin Clot Hypofibrinogenemia and factor XIII deficiency affect the
Lysis Time Test ELT. In hypofibrinogenemia, there is less fibrin to be lysed, and
The ELT is a global screen of the fibrinolytic system. In a short lysis time may be seen without a genuine increase in
the assay, plasma inhibitors of fibrinolysis are chemically fibrinolytic activity. In factor XIII deficiency, the original clot
removed, and the reactions of fibrinogen, plasminogen, and quality is poor, and digestion appears normal or speedy even
plasminogen activators are allowed to proceed uncontrolled. with low plasmin activity. The ELT also may be falsely pro-
Their effects are reported as a qualitative assessment using longed in chronic inflammation if the fibrinogen level exceeds
the ELT. 600mg/dL.
Patient plasma is diluted and acidified with cold 1% acetic
acid until the solution reaches a pH of 5.35 to 5.40. Upon
GLOBAL COAGULATION ASSAY:
refrigeration for approximately 30 minutes, a precipitate forms
THE THROMBOELASTOGRAPH
that contains fibrinogen, plasminogen, plasmin, and TPA. This
precipitate is termed the euglobulin fraction. Excluded from the The TEG Thromboelastograph Hemostasis Analyzer (Haemo-
precipitate are 2-antiplasmin and PAI-1; in their absence scope Corporation, Niles, Ill., a division of Haemonetics Cor-
fibrinolysis may proceed unchecked. The tubes are centrifuged, poration) is a global whole-blood analyzer that provides
and the supernatant is decanted completely. The precipitate is information on clotting dynamics, strength of clot, the effects
redissolved in borate buffer, and reagent thrombin or calcium of antithrombotics, the effects of platelets, and fibrinolysis. The
chloride is added. A clot should form within a few minutes. A thromboelastograph is used as a manual point-of-care coagu-
timer is started at the time of thrombin addition, and the clot lometer, mainly in cardiac surgery, and is described in detail in
is observed periodically for dissolution (disappearance) over 2 Chapter 47.

SUMMARY
Proper hemostasis specimen collection, transport, storage, and VWD is diagnosed and monitored through the judicious selection
centrifugation ensure a valid test result. and performance of platelet-based laboratory tests.
Specimens that are short draws, clotted, or hemolyzed are always The plasma markers PF4 and -thromboglobulin are used to
rejected. assess platelet activation.
Platelet aggregometry helps determine the cause of nonthrombo- Clot-based coagulation screening tests include the ACT, PT, PTT,
cytopenic mucocutaneous bleeding. and TCT.
CHAPTER 45 Laboratory Evaluation of Hemostasis 761

Mixing studies are used to detect factor deficiencies, LAs, and Tests of fibrinolysis include assays for FDPs, D-dimer, plasmino-
specific factor inhibitors. gen, TPA, and PAI-1.
Coagulation factor assays are used to detect and measure coagu-
lation factor deficiencies. Now that you have completed this chapter, go back and
Bethesda titers are used to detect and measure coagulation factor read again the case study at the beginning and respond
inhibitors. to the questions presented.

R E V I E W Q UESTIONS
1. What happens if a coagulation specimen collection tube 7. What is the gold standard assay for HIT?
is underfilled? a. Enzyme immunoassay
a. The specimen clots and is useless. b. SRA
b. The specimen is hemolyzed and is useless. c. Platelet lumiaggregometry
c. Clot-based test results are falsely prolonged. d. Washed platelet aggregation
d. Chromogenic test results are falsely decreased.
8. What agonist is used in platelet aggregometry to detect
2. If you collect blood into a series of tubes, when in the VWD?
sequence should the hemostasis (blue stopper) tube be a. Arachidonic acid
filled? b. Ristocetin
a. After a lavender-topped or green-topped tube c. Collagen
b. First, or after a nonadditive tube d. ADP
c. After a serum separator tube
d. Last 9. Deficiency of which single factor is likely when the PT
result is prolonged and the PTT result is normal?
3. What is the effect of hemolysis on a hemostasis specimen? a. Factor V
a. In vitro platelet and coagulation activation occur. b. Factor VII
b. The specimen is icteric or lipemic. c. Factor VIII
c. Hemolysis has no effect. d. Prothrombin
d. The specimen is clotted.
10. A prolonged PT, a low factor VII level, but a normal factor
4. Most coagulation testing must be performed on PPP, V level are characteristic of an acquired coagulopathy asso-
which is plasma with a platelet count less than: ciated with which of the following?
a. 1000/mcL a. Hemophilia
b. 10,000/mcL b. Liver disease
c. 100,000/mcL c. Thrombocytopenia
d. 1,000,000/mcL d. Vitamin K deficiency

5. You wish to obtain a 5-mL specimen of whole-blood/ 11. The patient has deep vein thrombosis. The PTT is pro-
anticoagulant mixture. The patients hematocrit is 65%. longed and is not corrected in an immediate mix of patient
What volume of anticoagulant should you use? plasma with an equal part of normal plasma. What is the
a. 0.32mL presumed condition?
b. 0.5mL a. Factor VIII inhibitor
c. 0.64mL b. LA
d. 0.68mL c. Factor VIII deficiency
d. Factor V Leiden mutation
6. You perform whole-blood lumiaggregometry on a speci-
men from a patient who complains of easy bruising. Aggre- 12. You perform a 5-mol/L urea solubility assay on a specimen
gation and secretion are diminished when the agonists from a patient with chronic bleeding and prolonged
thrombin, ADP, arachidonic acid, and collagen are used. wound healing. The clot dissolves within 2 hours. What is
What is the most likely platelet abnormality? the presumed cause?
a. Storage pool disorder a. Abnormally increased fibrinolysis
b. Aspirin-like syndrome b. Factor XIII deficiency
c. ADP receptor anomaly c. Factor IX deficiency
d. Glanzmann thrombasthenia d. The clot dissolution is normal
762 PART VIII Hemostasis and Thrombosis

13. What condition causes a pronounced elevation in the 15. What component of the fibrinolytic process binds and
result of the quantitative D-dimer assay? neutralizes free plasmin?
a. Deep vein thrombosis a. PAI-1
b. Fibrinogen deficiency b. TPA
c. Paraproteinemia c. 2-Antiplasmin
d. DIC d. Urokinase

14. What is the name given to the type of assay that uses a
synthetic polypeptide substrate which releases a chromo-
phore on digestion by its serine protease?
a. Clot-based assay
b. Molecular diagnostic assay
c. Fluorescence immunoassay
d. Chromogenic substrate assay

REFERENCES
1. Clinical and Laboratory Standards Institute: Collection, trans- 17. Horsti J: Use of EDTA samples for prothrombin time mea-
port, and processing of blood specimens for testing plasma-based surement in patients receiving oral anticoagulants. Haema-
coagulation assays: approved guideline, ed 5, CLSI Document tologica 86:851-855, 2001.
H21-A5, Wayne, Pa, 2008, Clinical and Laboratory Stan- 18. National Heart, Lung, and Blood Institute: The diagnosis,
dards Institute. evaluation, and management of von Willebrand disease, NIH
2. Clinical and Laboratory Standards Institute: Procedures for Publication No. 08-5832, Bethesda, Md, 2007, US Depart-
the collection of diagnostic blood specimens by venipuncture: ment of Health and Human Services, National Institutes of
approved standard, ed 6, CLSI Document H3-A6, Wayne, Pa, Health.
2007, Clinical and Laboratory Standards Institute. 19. Ens GE, Newlin F: Spurious protein S deficiency as a result
3. Clinical and Laboratory Standards Institute: Tubes and addi- of elevated factor VII levels. Clin Hemost Rev 9:18, 1995.
tives for venous blood specimen collection; approved standard, 20. Adcock D, Kressin D, Marlar RA: The effect of time and
ed 5, CLSI Document H1-A5, Wayne, Pa, 2007, Clinical and temperature variables on routine coagulation tests. Blood
Laboratory Standards Institute. Coagul Fibrinolysis 9:463-470, 1998.
4. Gottfried EL, Adachi MM: Prothrombin time and activated 21. Dyszkiewicz-Korpanty AM, Frenkel EP, Ravindra Sarode R:
partial thromboplastin time can be performed on the first Approach to the assessment of platelet function: comparison
tube. Am J Clin Pathol 107:681-683, 1997. between optical-based platelet-rich plasma and impedance-
5. Adcock DM, Kressin DC, Marlar RA: Minimum specimen based whole blood platelet aggregation methods. Clin Appl
volume requirements for routine coagulation testing. Am J Thromb Hemost 11:25-35, 2005.
Clin Pathol 109:595-599, 1998. 22. Pierangeli SS, Harris EN: Clinical laboratory testing for the
6. Ernst DJ: Blood specimen collection FAQs, Ramsey, Ind, 2008, antiphospholipid syndrome. Clin Chim Acta 357:17-33,
Center for Phlebotomy Education. 2005.
7. Marcus N: Coagulation testing in the context of the auto- 23. Ma Z, Monk TG, Goodnough LT, et al: Effect of hemoglobin-
mated hospital lab and optimizing for the effects of common and Perflubron-based oxygen carriers on common clinical
interferences. Clin Chem 56(suppl):A172, 2010 (abstract). laboratory tests. Clin Chem 43:1732-1737, 1997.
8. Marques M, Fritsma GA: Quick guide to coagulation testing, 24. Bick RL: Platelet function defects: A clinical review. Semin
ed 2, Washington, DC, 2009, AACC Press. Thromb Hemost 18:167-185, 1992.
9. Bennett A, Fritsma GA: Quick guide to venipuncture, 25. Dlott JS: Acquired platelet function disorders. In Goodnight
Washington, DC, 2010, AACC Press. SH, Hathaway WE, editors: Disorders of hemostasis and throm-
10. McCall R, Tankersley C: Phlebotomy essentials, ed 4, bosis, ed 2, New York, 2001, McGraw-Hill.
Philadelphia, 2007, Lippincott Williams & Wilkins. 26. Lind SE: The bleeding time does not predict surgical bleed-
11. Ernst C, Ernst DJ: Phlebotomy for nurses and nursing personnel, ing. Blood 77:2547-2552, 1991.
Ramsey, Ind, 2001, Center for Phlebotomy Education. 27. Duke WW: The pathogenesis of purpura haemorrhagica
12. Sunderji R, Gin K, Shalansky K, et al: Clinical impact of with especial reference to the part played by the blood plate-
point-of-care vs. laboratory measurement of anticoagula- lets. Arch Intern Med 10:445, 1912.
tion. Am J Clin Pathol 123:184-188, 2005. 28. Ivy AC, Nelson D, Bucher G: The standardization of certain
13. McGlasson DL, Paul J, Shaffer KM: Whole blood coagula- factors in the cutaneous venostasis bleeding time tech-
tion testing in neonates. Clin Lab Sci 6:76-77, 1993. nique. J Lab Clin Med 26:1812, 1941.
14. Clinical and Laboratory Standards Institute: Procedures and 29. Kumar R, Ansell JE, Canoso RT, et al: Clinical trial of a new
devices for the collection of diagnostic capillary blood specimens; bleeding device. Am J Clin Pathol 70:642-645, 1978.
approved standard, ed 6, CLSI Document H4-A6, Wayne, Pa, 30. Mielke CH, Jr, Kaneshiro MM, Maher IA, et al: The standard-
2008, Clinical and Laboratory Standards Institute. ized normal Ivy bleeding time and its prolongation by
15. Lippi G, Guidi GC: Effect of specimen collection on routine aspirin. Blood 34:204-215, 1969.
coagulation assays and D-dimer measurement. Clin Chem 31. Cattaneo M: Light transmission aggregometry and ATP
50:2150-2152, 2004. release for the diagnostic assessment of platelet function.
16. van den Besselaar AM, Meeuwisse-Braun J, Schaefer-van Semin Thromb Hemost 35:158-167, 2009.
Mansfeld H, et al: Influence of plasma volumetric errors on 32. McGlasson DL, Fritsma GA: Whole blood platelet
the prothrombin time ratio and international sensitivity aggregometry and platelet function testing. Semin Thromb
index. Blood Coagul Fibrinolysis 8:431-435, 1997. Hemost 35:168-180, 2009.
CHAPTER 45 Laboratory Evaluation of Hemostasis 763
33. Ingerman-Wojenski CM, Silver MJ: A quick method of after treatment of acute myocardial infarction with Alteplase.
screening platelet dysfunction using whole blood lumiag- Blood 78:1490-1495, 1991.
gregometry. Thromb Hemost 51:154-156, 1984. 52. Michelson AD: Laboratory markers of platelet activation. In
34. Goldenberg SJ, Veriabo NJ, Soslau G: A micromethod to Colman RW, Marder VJ, Clowes AW, et al, editors. Hemostasis
measure platelet aggregation and ATP release by impedance. and thrombosis: basic principles and clinical practice, ed 5,
Thromb Res 103:57-61, 2001. Philadelphia, 2006, Lippincott Williams & Wilkins, pp 825-
35. Dyszkiewicz-Korpanty AM, Frenkel EP, Sarode R: Approach 834.
to the assessment of platelet function: comparison between 53. Fritsma GA, Ens GE, Alvord MA, et al: Monitoring the anti-
optical-based platelet-rich plasma and impedance-based platelet action of aspirin. JAAPA 14:57-62, 2001.
whole blood platelet aggregation methods. Clin Appl Thromb 54. Eikelboom JW, Hankey GJ: Failure of aspirin to prevent
Hemost 11:25-35, 2005. atherothrombosis: potential mechanisms and implications
36. White MM, Foust JT, Mauer AM, et al: Assessment of lumiag- for clinical practice. Am J Cardiovasc Drugs 4:57-67, 2004.
gregometry for research and clinical laboratories. Thromb 55. Eikelboom JW, Hankey GJ, Thom J, et al: Incomplete inhibi-
Haemost 67:572-577, 1992. tion of thromboxane biosynthesis by acetylsalicylic acid:
37. Ren Q, Ye S, Whiteheart SW: The platelet release reaction: determinants and effect on cardiovascular risk. Circulation
just when you thought platelet secretion was simple. Curr 118:1705-1712, 2008.
Opin Hematol 15:537-541, 2008. 56. Lee RI, White PD: A clinical study of the coagulation time
38. Michiels JJ, Gadisseur A, Budde U, et al: Characterization, of blood. Am J Med Sci 243:279-285, 1913.
classification, and treatment of von Willebrand diseases: a 57. Clinical and Laboratory Standards Institute: One-stage pro-
critical appraisal of the literature and personal experiences. thrombin time (PT) test and activated partial thromboplastin
Semin Thromb Hemost 31:577-601, 2005. time (APTT) test; approved guideline, ed 2, CLSI Document
39. Budde U, Drewke E, Mainusch K, et al: Laboratory diagnosis H47-A2, Wayne, Penn, 2008, CLSI.
of congenital von Willebrand disease. Semin Thromb Hemost 58. Talstad I: Which coagulation factors interfere with the one-
28:173-190, 2002. stage prothrombin time? Haemostasis 23:19-25, 1993.
40. Allain JP, Cooper HA, Wagner RH, et al: Platelets fixed with 59. Biggs R, MacFarlane RG: Reaction of haemophilic plasma to
paraformaldehyde: a new reagent for assay of von Wille- thromboplastin. J Clin Pathol 4:445-459, 1951.
brand factor and platelet aggregating factor. J Lab Clin Med 60. Poller L: Laboratory control of anticoagulant therapy. Semin
85:318-328, 1975. Thromb Hemost 12:13-19, 1986.
41. Sharma RK, Reddy HK, Singh VN, et al: Aspirin and clopi- 61. Eisenberg JM, Clarke JR, Sussman SA: Prothrombin and
dogrel hyporesponsiveness and nonresponsiveness in partial thromboplastin times as preoperative screening tests.
patients with coronary artery stenting. Vasc Health Risk Arch Surg 117:48-51, 1982.
Manag 5:965-972, 2009. 62. Gravlee GP, Arora S, Lavender SW, et al: Predictive value of
42. Gu J, Prastein D, Pierson RN II: Aspirin resistance: an under- blood clotting tests in cardiac surgical patients. Ann Thorac
recognized risk factor in early vein graft thrombosis after Surg 58:216-221, 1994.
off-pump coronary artery bypass (OPCAB). Paper presented 63. McKinly L, Wrenn K: Are baseline prothrombin time/partial
at the American Heart Association Quality of Care and thromboplastin time values necessary before instituting
Outcomes Research in Cardiovascular Disease and Stroke anticoagulation? Ann Emerg Med 22:697-702, 1993.
Scientific Sessions, New Orleans, La., November 7-10, 64. Moll S, Ortel TL: Monitoring warfarin therapy in patients
2004. with lupus anticoagulants. Ann Intern Med 127:177-185,
43. Manoharan A, Brighton T, Gemmell R, et al: Platelet dys- 1997.
function in myelodysplastic syndromes: a clinicopathologi- 65. McGlasson DL, Romick BG, Rubal BJ: Comparison of a
cal study. Int J Hematol 76:272-278, 2002. chromogenic factor X assay with international normalized
44. Brace LD: Testing for heparin-induced thrombocytopenia by ratio for monitoring oral anticoagulation therapy. Blood
platelet aggregometry. Clin Lab Sci 5:80-81, 1992. Coagul Fibrinolysis 19:513-517, 2008.
45. Warkentin T: Heparin-induced thrombocytopenia. In 66. Lusher JM: Screening and diagnosis of coagulation disor-
Colman RW, Marder VJ, Clowes AW, et al, editors: Hemostasis ders. Am J Obstet Gynecol 175:778-783, 1996.
and thrombosis: basic principles and clinical practice, ed 5, 67. Sibley C, Singer JW, Wood RJ: Comparison of activated
Philadelphia, 2006, Lippincott Williams & Wilkins, pp 1649- partial thromboplastin reagents. Am J Clin Pathol 59:581-
1662. 586, 1973.
46. Stewart MW, Etches WS, Boshkov LK, et al: Heparin-induced 68. Triplett DA, Harms CS, Koepke JA: The effect of heparin
thrombocytopenia: an improved method of detection based on the partial thromboplastin time. Am J Clin Pathol
on lumi-aggregometry. Br J Haematol 91:173-177, 1995. 70(suppl):556-559, 1978.
47. Amiral J, Bridey F, Dreyfus M, et al: Platelet factor 4 com- 69. Brill-Edwards P, Ginsberg JS, Johnston M, et al: Establishing
plexed to heparin is the target for antibodies generated in a therapeutic range for heparin therapy. Ann Intern Med
heparin-induced thrombocytopenia. Thromb Hemost 68:95- 119:104-109, 1993.
96, 1992. 70. Hathaway WE, Assmus SL, Montgomery RR, et al: Activated
48. Sadayasu T, Nakashima Y, Yashiro A, et al: Heparin-releasable partial thromboplastin time and minor coagulopathies. Am
platelet factor 4 in patients with coronary artery disease. Clin J Clin Pathol 71:22-25, 1979.
Cardiol 14:725-729, 1991. 71. McKinly L, Wrenn K: Are baseline prothrombin time/partial
49. Rand ML, Leung R, Packham MA: Platelet function assays. thromboplastin time values necessary before instituting
Transfus Apher Sci 28:307-317, 2003. anticoagulation? Ann Emerg Med 22:697-702, 1993.
50. Darling CE, Michelson AD, Volturo GA, et al: Platelet reac- 72. McNeil HP, Simpson RJ, Chesterman CN, et al: Antiphos-
tivity and the identification of acute coronary syndromes in pholipid antibodies are directed against a complex antigen
the emergency department. J Thromb Thrombolysis 28:31-37, that includes a lipid-binding inhibitor of coagulation: -2-
2009. glycoprotein I. Proc Natl Acad Sci U S A 87:4120-4124, 1990.
51. Rapold HJ, Grimaudo V, Declerck PJ, et al: Plasma 73. Ledford-Kraemer MR: Laboratory testing for lupus anti
levels of plasminogen activator inhibitor type 1, beta- coagulants: pre-examination variables, mixing studies, and
thromboglobulin, and fibrinopeptide A before, during, and diagnostic criteria. Semin Thromb Hemost 34:380-388, 2008.
764 PART VIII Hemostasis and Thrombosis

74. Pengo V, Tripodi A, Reber G: Update of the guidelines for 94. Stein PD, Hull RD, Patel KC, et al: D-dimer for the exclusion
lupus anticoagulant detection. Subcommittee on Lupus of acute venous thrombosis and pulmonary embolism: a
Anticoagulant/Antiphospholipid Antibody of the Scientific systematic review. Ann Intern Med 140:589-602, 2004.
and Standardisation Committee of the International Society 95. Kline JA, Israel EG, Michelson EA: Diagnostic accuracy of a
on Thrombosis and Haemostasis. J Thromb Haemost 7:1737- bedside D-dimer assay and alveolar dead-space measure-
1740, 2009. ment for rapid exclusion of pulmonary embolism: a multi-
75. Eika C, Godal HC, Kierulf P: Detection of small amounts of center study. JAMA 285:761-768, 2001.
heparin by the thrombin clotting-time. Lancet 300:376-377, 96. Drewinko B, Surgeon J, Cobb P, et al: Comparative sensitiv-
1972. ity of different methods to detect and quantify circulating
76. Meh DA, Siebenlist KR, Bergtrom G, et al: Sequence of fibrinogen/fibrin split products. Am J Clin Pathol 84:58-66,
release of fibrinopeptide A from fibrinogen molecules by 1985.
thrombin or Atroxin. J Lab Clin Med 125:384-391, 1995. 97. Garvey MB, Black JM: The detection of fibrinogen-fibrin
77. Clinical and Laboratory Standards Institute: Procedure for the degradation products by means of a new antibody-coated
determination of fibrinogen in plasma: approved guideline, ed 2, latex particle. J Clin Pathol 25:680-682, 1972.
CLSI Document H30-A2, Wayne, Pa, 2001, Clinical and 98. Lottenberg R, Dolly FR, Kitchens CS: Recurring thromboem-
Laboratory Standards Institute. bolic disease and pulmonary hypertension associated with
78. Poller L, Thomson JM, Yee KF: Fibrinogen determination in severe hypoplasminogenemia. Am J Hematol 19:181-193,
a series of proficiency studies. Clin Lab Haematol 2:43-51, 1985.
1980. 99. Walenga JM: Molecular and automated assessments of coag-
79. Comp PC: Overview of the hypercoagulable states. Semin ulation. In Corriveau DM, Fritsma GA, editors: Hemostasis
Thromb Hemost 16:158-161, 1990. and thrombosis in the clinical laboratory, Philadelphia, 1988,
80. Seligsohn U: High gene frequency of factor XI (PTA) defi- JB Lippincott.
ciency in Ashkenazi Jews. Blood 51:1223-1228, 1978. 100. Friberger P: Synthetic peptide substrate assays in coagula-
81. Rothschild C, Amiral J, Adam M, et al: Preparation of factor tion and fibrinolysis and their application on automates.
VIII depleted plasma with antibodies and its use for the Semin Thromb Hemost 9:281-300, 1983.
assay of factor VIII. Haemostasis 20:321-328, 1990. 101. Bovill EG: Fibrinolytic defects and thrombosis. In Hathaway
82. Arkin CF, Bovill EG, Brandt JT, et al: Factors affecting the WE, Goodnight SH, editors: Disorders of hemostasis and
performance of factor VIII coagulant activity assays: results thrombosis: a clinical guide, ed 2, New York, 2001, McGraw-
of proficiency surveys of the College of American Patholo- Hill, pp 389-398.
gists. Arch Pathol Lab Med 116:908-915, 1992. 102. Booth NA, Bachmann F: Plasminogen-plasmin system. In
83. Hirst CF, Hewitt J, Poller L: Trace levels of factor VIII in Colman RW, Marder VJ, Clowes AW, et al, editors: Hemostasis
substrate plasma. Br J Haematol 81:305-306, 1992. and thrombosis: basic principles and clinical practice, ed 5,
84. Kershaw G, Jayakodi D, Dunkley S: Laboratory identifica- Philadelphia, 2006, Lippincott Williams & Wilkins, pp 335-
tion of factor inhibitors: the perspective of a large tertiary 380.
hemophilia center. Semin Thromb Hemost 35:760-768, 2009. 103. Wiman B, Hamsten A: The fibrinolytic enzyme system and
85. Greenberg CS, Sane DC, Lai T: Factor XIII and fibrin stabi- its role in the etiology of thromboembolic disease. Semin
lization. In Colman RW, Marder VJ, Clowes AW, et al, Thromb Hemost 16:207-216, 1990.
editors: Hemostasis and thrombosis: basic principles and clinical 104. Patrassi GM, Sartori MT, Casonato A, et al: Urokinase-type
practice, ed 5, Philadelphia, 2006, Lippincott Williams & plasminogen activator release after DDAVP in von Wille-
Wilkins, pp 317-334. brand disease: different behaviour of plasminogen activa-
86. Krumdieck R, Shaw DR, Huang ST, et al: Hemorrhagic dis- tors according to the synthesis of von Willebrand factor.
order due to an isoniazid-associated acquired factor XIII Thromb Res 66:517-526, 1992.
inhibitor in a patient with Waldenstrms macroglobulin- 105. Cushman M, Alving BM: Risk factors for cardiovascular
emia. Am J Med 90:639-645, 1991. disease and arterial thrombosis. In Kitchens CA, Alving BM,
87. Karimi M, Bereczky Z, Cohan N, et al: Factor XIII deficiency. Kessler CM: Consultative hemostasis and thrombosis, ed 2,
Semin Thromb Hemost 35:426-438, 2009. Philadelphia, 2007, Saunders, pp 371-378.
88. Southern DK: Serum FDP and plasma D-dimer testing: what 106. Nicholl SM, Roztocil E, Davies MG: Plasminogen activator
are they measuring? Clin Lab Sci 5:332-333, 1992. system and vascular disease. Curr Vasc Pharmacol 4:101-116,
89. Lawler CM, Bovill EG, Stump DC, et al: Fibrin fragment 2006.
D-dimer and fibrinogen B beta peptides in plasma as 107. Chandler WL, Schmer G, Stratton JR: Optimum conditions
markers of clot lysis during thrombolytic therapy in acute for the stabilization and measurement of tissue plasmino-
myocardial infarction. Blood 76:1341-1348, 1990. gen activator activity in human plasma. J Lab Clin Med
90. Waser G, Kathriner S, Wuillemin WA: Performance of the 113:362-371, 1989.
automated and rapid STA Liatest D-dimer on the STA-R 108. Juhan-Vague I, Alessi MC, Raccah D, et al: Daytime fluctua-
analyzer. Thromb Res 116:165-170, 2005. tion of plasminogen activator inhibitor 1 (PAI-1) in popula-
91. Reber G, Bounameaux H, Perrier A, et al: Evaluation of tions with high PAI-1 levels. Thromb Haemost 67:76-82, 1992.
advanced D-dimer assay for the exclusion of venous throm- 109. Macy EM, Meilahn EN, Declerck PJ, et al: Sample prepara-
boembolism. Thromb Res 107:197-200, 2002. tion for plasma measurement of plasminogen activator
92. Sadanaga T, Sadanaga M, Ogawa S: Evidence that D-dimer inhibitor-1 antigen in large population studies. Arch Pathol
levels predict subsequent thromboembolic and cardio Lab Med 117:67-70, 1993.
vascular events in patients with atrial fibrillation during 110. Philips M, Juul A, Selmer J, et al: A specific immunologic
oral anticoagulant therapy. J Am Coll Cardiol 55:2225-2231, assay for functional plasminogen activator inhibitor 1 in
2010. plasma standardized measurements of the inhibitor and
93. Heijboer H, Ginsberg JS, Bller HR, et al: The use of the related parameters in patients with venous thromboembolic
D-dimer test in conjunction with noninvasive testing vs. disease. Thromb Haemost 68:486-494, 1992.
serial noninvasive testing alone for the diagnosis of deep 111. Vinazzer H: Basics and practice in evaluating plasminogen.
vein thrombosis. Thromb Haemost 67:510-513, 1992. Haemostasis 18(suppl 1):41-45, 1988.
Monitoring Antithrombotic Therapies
George A. Fritsma
46
OUTLINE OBJECTIVES
Warfarin Therapy and the After completion of this chapter, the reader will be able to:
Prothrombin Time
Warfarin: Vitamin K 1. Describe the purpose of antithrombotic administra- 8. Perform and interpret the results of the chromogenic
Antagonist tion and distinguish between anticoagulants and anti-Xa assay for the monitoring of unfractionated
Warfarin Prophylaxis and antiplatelet therapy. heparin therapy, low-molecular-weight heparin
Therapy 2. Describe the indications for, dosage of, and manage- therapy, and fondaparinux therapy.
Monitoring Warfarin ment of warfarin therapy, including how to treat 9. Select appropriate laboratory tests for the monitoring
Therapy Using the warfarin overdose. of antithrombotic therapy, recognize appropriate
Prothrombin Time Assay 3. Monitor warfarin therapy using the prothrombin time testing frequency, follow appropriate timing for test
Monitoring Warfarin and international normalized ratio, and compare administration, and interpret test results as indicat-
Therapy Using the these tests with the chromogenic X assay. ing adequate, inadequate, or excessive drug dosage.
Chromogenic Factor X
4. Perform and interpret prothrombin times using point- 10. Describe how rivaroxaban exerts its direct anti-Xa
Assay
of-care instruments. effect; discuss methods for monitoring rivaroxaban
Effect of Diet and Drugs
on Warfarin Therapy 5. Describe the indications for, dosage of, and manage- therapy.
Effect of Polymorphisms ment of unfractionated heparin therapy, including 11. Monitor therapy with direct thrombin inhibitors,
on Warfarin Therapy how to establish the unfractionated heparin thera- including dabigatran, using the partial thromboplas-
Effect of Direct Thrombin peutic partial thromboplastin time range. tin time and the ecarin clotting time.
Inhibitors on the 6. Describe the indications, dosage, and management 12. Describe the therapeutic function of the intravenous
Prothrombin Time of low-molecular-weight heparin therapy and platelet glycoprotein IIb/IIIa inhibitors and describe
Reversal of Warfarin fondaparinux therapy. how their effects are monitored.
Overdose 7. Perform and interpret the results of the partial throm- 13. Describe the therapeutic function of the oral platelet
Prothrombin Time boplastin time and activated clotting time assays for inhibitors aspirin, clopidogrel, ticlopidine, and prasu-
Point-of-Care Testing
the monitoring of unfractionated heparin therapy, grel and describe how their effects are monitored.
Unfractionated Heparin
and compare these tests with the chromogenic
Therapy and the Partial
Thromboplastin Time anti-Xa heparin assay.
Heparin Is a Catalyst That
Activates Antithrombin to
Neutralize Serine Proteases
Unfractionated Heparin
Therapy
Monitoring Unfractionated
Heparin Therapy Using
the Partial CASE STUDY
Thromboplastin Time After studying the material in this chapter, the reader should be able to respond to the following
Determining the Partial case study:
Thromboplastin Time
Therapeutic Range for A 71-year-old woman with atrial fibrillation has been taking 5mg of warfarin (Coumadin) per day
Unfractionated Heparin for 8 years. Her monthly prothrombin time international normalized ratio (INR) has been main-
Therapy tained consistently within the therapeutic range, 2 to 3. Last Monday, however, her prothrombin
Clinical Utility of Partial time INR was 6.5. For several days earlier she had noticed that her gums bled after she brushed
Thromboplastin Time her teeth.
Monitoring of
1. What could cause this change in the prothrombin time (INR) result?
Unfractionated Heparin
2. What should be done about her INR and symptoms?
Therapy and Reporting
of Results 3. What alternative test may be used to monitor warfarin therapy?

765
766 PART VIII Hemostasis and Thrombosis

V
enous or arterial thrombotic disease is managed by antithrombotic therapy, which includes
OUTLINEcontd
anticoagulant and antiplatelet therapy. Venous thromboembolic disease (venous thromboembolism,
Limitations of Partial or VTE) includes superficial and deep vein thrombosis (DVT) and pulmonary embolism (PE) and
Thromboplastin is treated using the anticoagulants warfarin (Coumadin), standard unfractionated heparin (UFH),
Time Monitoring of low-molecular-weight heparin (LMWH, enoxaparin), and synthetic pentasaccharide (fondaparinux).
Unfractionated Heparin The direct thrombin inhibitors (DTIs) argatroban, lepirudin, and bivalirudin are additional anticoagulants
Therapy
substituted for heparin in patients who have developed heparin-induced thrombocytopenia (HIT) sub
Monitoring Unfractionated
sequent to UFH therapy (see Chapter 42). All of these anticoagulant drugs are administered intra
Heparin Therapy Using
the Activated Clotting venously except warfarin, which is an oral anticoagulant. Unfractionated heparin therapy began in the
Time late 1930s, and warfarin therapy was first used in 1952. As of July 2010, warfarin remained the only
Reversal of Unfractionated oral anticoagulant, although at least two new oral anticoagulants, rivaroxaban and dabigatran, may be
Heparin Overdose Using cleared by the U.S. Food and Drug Administration (FDA) in 2011 or 2012.
Protamine Sulfate Arterial thrombosis includes acute myocardial infarction (AMI), ischemic cerebrovascular accident
Low-Molecular-Weight (stroke), transient ischemic attack, and peripheral arterial occlusion and is managed with the heparins, the
Heparin Therapy and DTIs, and the antiplatelet drugs aspirin, ticlopidine, and clopidogrel. Aspirin is particularly effective
the Chromogenic Anti in reducing mortality when taken within 6 hours of the onset of symptoms. The platelet glycoprotein
Factor Xa Heparin Assay IIb/IIIa inhibitor drugs eptifibatide, abciximab, and tirofiban are used to prevent thrombosis during
Low-Molecular-Weight
cardiac catheterization procedures.
Heparin Is Derived from
Thrombolytic therapy helps resolve DVT, PE, peripheral artery occlusions, AMI, and stroke, particularly
Unfractionated Heparin
Monitoring Low-Molecular- when used shortly after the onset of symptoms. Most thrombolytic therapy uses the recombinant
Weight Heparin Therapy forms of tissue plasminogen activator, reteplase and alteplase. Thrombolytic therapy raises the risk of
Monitoring hemorrhage, particularly intracranial hemorrhage. Thrombolytic therapy is not monitored by labor
Pentasaccharide atory tests.
Therapy Using the Many lives have been saved through judicious use of antithrombotic therapy, and countless
Chromogenic Anti more healthy individuals have been spared thrombotic disease through long-term antithrombotic
Factor Xa Heparin Assay prophylaxis. However, antithrombotics are dangerous because their effective dosage ranges are
Rivaroxaban, an Oral narrow.1 Overdose is critical and leads to emergency department visits for uncontrolled bleeding;
Direct Factor Xa Inhibitor inadequate dosages lead to secondary (repeat), often fatal, thrombotic events. Dosages and half-lives
Monitoring Direct Thrombin
differ among the antithrombotics and thrombolytics because of variations in formulation and
Inhibitor Therapy
metabolism.2,3
Using the Partial
Thromboplastin Time or Because of these risks, laboratory monitoring of antithrombotic therapy, the earliest form of thera-
Ecarin Clotting Time peutic drug monitoring, is essential. Coagulation laboratory scientists perform countless prothrombin
Argatroban time (PT) assays, partial thromboplastin time (PTT, activated partial thromboplastin time [APTT])
Lepirudin, a Recombinant assays, and chromogenic antifactor Xa heparin assays to monitor anticoagulant therapy, and this accounts
Analogue of Hirudin for most of their workload. Antiplatelet therapy monitoring generates a smaller, but rapidly growing
Bivalirudin, a Recombinant workload. Although antithrombotic therapy monitoring may seem routine, vigilance is essential to
Derivative of Hirudin provide consistently valid results in a dangerous therapeutic world.4
Dabigatran, an Oral Direct
Thrombin Inhibitor
Monitoring Direct Thrombin WARFARIN THERAPY AND THE PROTHROMBIN TIME
Inhibitor Therapy
Monitoring Antiplatelet
Warfarin: Vitamin K Antagonist
Therapy Using Platelet As detailed in Chapter 40, coagulation factors II (prothrombin), VII, IX, and X depend on vitamin K
Activity Assays for normal production, as do coagulation control proteins C, S, and Z. Vitamin K is responsible for
Intravenous Glycoprotein -carboxylation of a series of 12 to 18 N-terminus glutamic acids, a posttranslational modification that
IIb/IIIa Inhibitors Are enables these coagulation factors and control proteins to bind ionic calcium (Ca2+) and cell membrane
Used During Cardiac phospholipids, especially phosphatidylserine (see Figure 40-8). Vitamin K is concentrated in green
Catheterization leafy vegetables and is produced by gut flora; its absence results in the production of nonfunctional
Aspirin, Clopidogrel, des- carboxyl, forms of factors II, VII, IX, and X and proteins C, S, and Z.
Ticlopidine, and Prasugrel Warfarin sodium (4-hydroxycoumarin, Coumadin) is a member of the coumarin drug family and
Reduce the Incidence of is the formulation of coumarin most often used in North America.5 Another coumarin is dicumarol
Arterial Thrombosis
(3,3-methylenebis-[4-hydroxycoumarin]), the original anticoagulant extracted from moldy sweet
Variable Antiplatelet
clover by Link in 1940.6 Warfarin is a vitamin K antagonist that suppresses -carboxylation of glutamic
Response and
Laboratory Monitoring of acid by slowing the activity of the enzyme vitamin K epoxide reductase (see Figure 40-8). During warfarin
Antiplatelet Resistance therapy, the activities of factors II, VII, IX, and X and proteins C, S, and Z become reduced as the
Future of Antithrombotic nonfunctional des-carboxyl proteins are produced. These are called proteins induced by vitamin K antago-
Therapy nists (PIVKAs); they bind few calcium ions, do not assemble on phospholipid surfaces with their
substrates, and therefore do not participate in coagulation. Despite several developmental efforts, in
CHAPTER 46 Monitoring Antithrombotic Therapies 767

100 TABLE 46-1 Plasma Half-Life, Normal Plasma Level,


and Minimum Effective Hemostatic Level as a Percentage of
80 Normal Level for the Coagulation Factors
Percent activity

60 Factor Half-Life Plasma Level Hemostatic Level

40 Fibrinogen 4 days 280mg/dL 50mg/dL


Prothrombin Prothrombin 60hr 1300mcg/mL 20%
20 X
IX V 16hr 680mcg/mL 25%
VII VII 6hr 120mcg/mL 20%
1 2 3 4 5 VIII 12hr 0.24mcg/mL 30%
Days of warfarin therapy IX 24hr 5mcg/mL 30%
Figure 46-1 Factor VII activity decreases to 50% of normal 6 hours after X 30hr 1mg/dL 25%
warfarin therapy is begun, prolonging the factor VIIasensitive prothrombin XI 2-3 days 6mcg/mL 25%
time to near the therapeutic range. The half-lives of factors II (prothrombin), XIII 7-10 days 290mcg/mL 2-3%
IX, and X are longer than that of VII; factor II activity requires at least 3 days Von Willebrand 30hr 6mcg/mL 50%
to decline by 50%. The patient gains full anticoagulation effects approxi- factor
mately 5 days after the start of warfarin therapy.

2010 coumarins remained the only oral anticoagulants in the Monitoring Warfarin Therapy Using
United States, so warfarin therapy is often called oral anticoagu- the Prothrombin Time Assay
lant therapy. Two new oral anticoagulant drugs, rivaroxaban and The PT effectively monitors warfarin therapy because it is sensi-
dabigatran, became available in 2009 outside the United States tive to reductions of factors II, VII, and X (Figure 46-2). The PT
and may be cleared by the FDA in 2011 or 2012. reagent consists of tissue factor, phospholipid, and ionic
calcium, so it triggers the coagulation pathway at the level of
Warfarin Prophylaxis and Therapy factor VII. Owing to the 6-hour half-life of factor VII, the PT
Warfarin is prescribed prophylactically to prevent strokes in begins to prolong within 6 to 8 hours; however, anticoagulation
patients with atrial fibrillation and to prevent VTE after trauma, becomes therapeutic only when the activities of factors II and
orthopedic surgery, or general surgery, and in a number of X decrease to less than 50% of normal, in approximately 5 days.
chronic medical conditions. Therapeutic warfarin inhibits the The first PT is assayed 24 hours after therapy is initiated;
recurrence of DVT or PE. Warfarin is also used therapeutically subsequent PTs are performed daily until two consecutive
after AMI if the event is complicated by congestive heart failure results are within the target therapeutic range. Monitoring con-
or coronary insufficiency. Warfarin is among the top 20 most tinues every 4 weeks until the completion of therapy, which
commonly prescribed drugs in North America. often lasts for 6 months following a thrombotic event. Warfarin
The standard warfarin regimen begins with a 5-mg daily oral therapy for atrial fibrillation is of indefinite length, possibly
dose. The starting dosage for people older than 70 years and lifelong. Close monitoring is essential for successful warfarin
people who are debilitated, malnourished, or have congestive therapy, because the therapeutic range is narrow. Underantico-
heart failure is 2mg/day. For people simultaneously taking agulation signals the danger of thrombosis or secondary
drugs that are known to raise warfarin sensitivity, the starting thrombosis (rethrombosis); overdose carries the danger of
dosage is 2mg/day, and 2mg/day is also the dosage used for hemorrhage.
those with inherited warfarin sensitivity. There is no loading
dose, and subsequent dosing is based on patient response as Reporting Prothrombin Time Results and
measured by the PT (see the next section). The activity of each the International Normalized Ratio
of the vitamin Kdependent coagulation factors begins to The medical laboratory scientist reports PT results to the nearest
decline immediately but at different rates (Figure 46-1) for tenth of a second and provides the PT reference interval for
each factor, and it takes about 5 days for all the factors to reach comparison. In view of the inherent variations among throm-
therapeutic levels. Table 46-1 lists the plasma half-life, plasma boplastin reagents, and to accomplish interlaboratory normal-
concentration, and minimum effective plasma percentage of ization, all laboratories report the international normalized
normal factor activity for the coagulation factors. ratio (INR) for patients with a stable response to anticoagulant
Control protein activities also become reduced, especially therapy using the following formula8:
the activity of protein C, which has a 6-hour half-life, so for
the first 2 or 3 days of warfarin therapy the patient actually risks INR = (PTpatient PTnormal )ISI
a thrombotic event. For this reason, warfarin therapy is
covered by UFH, LMWH, or pentasaccharide therapy for at where PTpatient is the PT of the patient in seconds, PTnormal is the
least 5 days. Failure to provide anticoagulant therapy during geometric mean of the PT reference interval in seconds, and ISI
this period may result in warfarin skin necrosis, a severe reaction is the international sensitivity index applied as an exponent in
requiring dbridement of dead tissue.7 the formula.
768 PART VIII Hemostasis and Thrombosis

Vlla Xla Xlla

TF Pre-K HMWK

IXa Vllla PTT reagent: negatively


charged particulate
activator, phospholipid,
Ca2; prolonged by
PT reagent: tissue factor, Xa Va deficiencies of XII, XI, IX,
phospholipid, Ca2; PT is VIII, X, V, II, fibrinogen
prolonged by deficiencies of
VII, X, V, II, fibrinogen
Thr Xllla

Cross-linked
Fibrinogen Fibrin polymer
fibrin

Figure 46-2 The prothrombin time (PT) reagent activates the extrinsic coagulation pathway beginning with factor VII. The PT is prolonged by deficiencies
of factors VII, X, V, II (prothrombin), and fibrinogen when the fibrinogen concentration is less than 100mg/dL. The PT is prolonged in warfarin therapy, because
it responds to the reduced VII, X, and prothrombin activity. The partial thromboplastin time (PTT) reagent activates the intrinsic coagulation pathway through
factor XII, assisted by prekallikrein (pre-K) and high-molecular-weight kininogen (HMWK). The PTT is prolonged by deficiencies of pre-K; HMWK; factors XII,
XI, IX, VIII, and X; prothrombin; and fibrinogen when the fibrinogen concentration is less than 100mg/dL. The PTT is prolonged in unfractionated heparin
(UFH) therapy because UFH activates the plasma control protein antithrombin, which neutralizes the serine proteases XIIa, XIa, Xa, IXa, and IIa (thrombin [Thr]).
TF, Tissue factor.

Thromboplastin producers generate the ISI by performing Monitoring Warfarin Therapy Using
an orthogonal regression analysis comparing the results of the Chromogenic Factor X Assay
their PT reagents for 50 or more anticoagulated specimens The chromogenic coagulation factor X assay may be used as an
and 10 or more normal specimens with the published results alternative to the PT/INR system, eliminating the necessity for
of the international reference thromboplastin (World Health normalization. The therapeutic range is determined locally and
Organization human brain thromboplastin).9 Most manufac- typically is close to 20% to 40% of normal factor X activity.
turers provide ISIs for a variety of coagulation instruments, The chromogenic X assay is useful when the PT is compromised
because each may respond differently to their thromboplas- by an acquired or congenital coagulation factor deficiency.
tins; for instance, some coagulometers rely on photometric
changes, whereas others use an electromechanical system (see Effect of Diet and Drugs on Warfarin Therapy
Chapter 47). Most thromboplastin reagents have ISIs near 1, Dietary vitamin K decreases warfarins effect and reduces the
which is the ISI of the World Health Organizations throm- INR. Green vegetables are an important source of vitamin K,
boplastin. Automated coagulation timers request the reagent but vitamin K also is concentrated in liver, avocados, parenteral
ISI from the operator or obtain it directly from a reagent nutrition formulations, multivitamins, red wine, over-the-
vial bar code and compute the INR for each assay result. counter nutrition drinks, and over-the-counter dietary supple-
Although INRs are meant to be computed only for patients ments. A patient who is taking warfarin is counseled to maintain
taking anticoagulants in whom the response to therapy has a regular balanced diet and to follow up dietary or dietary
stabilized, they typically are reported for all patients, even supplement changes with additional PT assays and dosage
those who are not taking warfarin. During the first 5 days adjustments if necessary.
of warfarin therapy, the astute physician and medical labora- Warfarin is metabolized in the mitochondrial cytochrome
tory scientist ignore the INR as unreliable and interpret P-450 (CYP 2C9) pathway of hepatocytes, the disposal system
the PT results in seconds, comparing it with the reference for at least 80 drugs. Theoretically, use of any drug metabolized
interval. through the CYP 2C9 pathway may unpredictably suppress or
enhance the effects of warfarin. Amiodarone, metronidazole,
Warfarin International Normalized Ratio and cimetidine typically double or triple the INR. Any change
Therapeutic Range in drug therapy, like a change in diet, must be followed up with
The physician adjusts the warfarin dosage to achieve the desired additional PT assays and dosage adjustments.
INR of 2 to 3, or 2.5 to 3.5 if the patient has a mechanical heart Warfarin is contraindicated during pregnancy, because it
valve. INRs greater than 4 are associated with increased risk of causes birth defects. When anticoagulation is desired during
hemorrhage, and such a finding requires immediate commu- pregnancyfor instance, in women who possess a thrombosis
nication with the clinician who is managing the patients case. risk factorLMWH or fondaparinux is prescribed.
Adjustments are made conservatively because the INR requires
4 to 7 days to stabilize at the new target level, but an elevated Effect of Polymorphisms on Warfarin Therapy
INR accompanied by the symptoms of anatomic bleeding is a Two polymorphisms generate variations in enzymes of the cyto
medical emergency. chrome P-450 pathway. These are CYP2C9*2 and CYP2C9*3,
CHAPTER 46 Monitoring Antithrombotic Therapies 769
which reduce pathway activity and slow the metabolic break- TABLE 46-2 Recommendations for the Reversal of
down of warfarin. Likewise, there is a polymorphism that affects Warfarin Overdose Based on International Normalized Ratio
the key enzyme of vitamin K metabolism, vitamin K epoxide (INR) and Bleeding
reductase. This polymorphism, named VKORC1, slows vitamin
Bleeding INR Intervention
K reduction, which makes the patient more sensitive to warfa-
rin. In patients possessing one, two, or all three of these poly- No significant 3-5 Reduce dosage or omit one dose,
morphisms, warfarin therapy should begin at 2mg/day and bleeding monitor INR frequently
should be adjusted and monitored daily until the INR remains 5-9 Omit warfarin, monitor INR frequently,
consistently in the therapeutic range. The standard 5mg/day consider oral vitamin K (5mg) if
regimen risks hemorrhage in these patients. In 2007, the FDA high risk for bleeding (surgery)
required that drug manufacturers add a statement on all vials
>9 Stop warfarin, give 5-10mg oral
of warfarin recommending that physicians screen patients for
vitamin K, monitor INR frequently
dosage-affecting polymorphisms. Although the FDA recom-
Serious Any INR Stop warfarin; give 10mg vitamin K by
mendation does not carry the weight of a black box warning,
bleeding intravenous push, may repeat every
numerous molecular diagnostics manufacturers have devel-
12hr; give thawed frozen plasma,
oped short turn-around assays for these three polymorphisms.
prothrombin complex concentrate,
Screening for these polymorphisms is the first and most public
or recombinant factor VIIa
example of pharmacogenomic laboratory testing, although as
Life-threatening Any INR Same as for serious bleeding, except
of 2010 it had not been universally endorsed.10
bleeding stronger indication for recombinant
Conversely, warfarin receptor insufficiency may render the
factor VIIa
patient resistant to warfarin therapy. Some patients require
dosages of 20mg/day or higher to achieve a therapeutic INR.
The search is on for polymorphisms responsible for such war-
farin resistance, and by 2012 or 2013, at least one such assay HMS Plus, ACT Plus (bench top, Medtronic Cardiac Surgery,
may become available from reference laboratories.11 Minneapolis, Minn.)
INRatio PT INR monitor, INRatio2 PT INR monitor
Effect of Direct Thrombin Inhibitors (HemoSense/Inverness Medical, San Diego, Calif.)
on the Prothrombin Time i-STAT 1 (Abbott Point of Care, Inc., Princeton, N.J.)
The intravenously administered DTIs argatroban, lepirudin, ProTime Microagulation SystemNew ProTime; Hemo-
and bivalirudin, which are used in place of heparin as a life- chron Signature Elite; Hemochron Signature Plus; Hemo-
saving measure for patients with HIT, prolong the PT, although chron Response (International Technidyne Corporation,
the PTT is the assay typically employed to monitor DTI therapy. Edison, N.J.)
In subsequent switching from DTI to warfarin therapy, the Most of these instruments are portable and are hand-held.13
clinician is aware that the combination of a DTI and warfarin Point-of-care instruments permit near-patient PT testing at
nearly doubles the PT for the duration of action of the DTI, anticoagulation clinics, bedside testing, self-testing, and testing
which is 3 or 4 days. Argatroban exerts the greatest effect on of infants. The operator or patient performs a capillary punc-
the PT, followed by bivalirudin and lepirudin.12 The chromo- ture and captures the free-flowing capillary blood in a cartridge
genic X assay is an effective means for monitoring warfarin that is transferred to the instrument. Alternatively, a drop of
dosage during the crossover period. anticoagulated venous whole blood may be transferred to the
test cartridge. No point-of-care instrument measures PTT,
Reversal of Warfarin Overdose although several report activated clotting time (ACT).
Table 46-2 provides recommendations for the reversal of a Each instrument uses individual patented technology to
warfarin overdose based on INR and clinical evidence of bleed- generate a PT with an INR, and each produces reference inter-
ing. The concerns surrounding warfarin therapy will recede in vals and target therapeutic ranges in INR units and seconds that
importance as new oral anticoagulants are cleared for distribu- may diverge slightly from plasma-based central laboratory
tion in the United States (see the sections on rivaroxaban and results. In many cases, the manufacturer programs in a linear
dabigatran). electronic conversion factor so that the readout more closely
matches plasma results. The manufacturer also provides inter-
Prothrombin Time Point-of-Care Testing nal electronic calibration and whole-blood controls. Before
The following instruments have been cleared by the FDA for point-of-care instruments are placed in service, laboratory
point-of-care PT measurement using a venous whole-blood scientists validate the units measurements using a plasma-
specimen or capillary specimen, or both: based PT assay as the reference method, comparing the results
Cascade POC, Actalyke XL and Actalyke Mini II (Helena obtained by each method through linear regression and a test
Point of Care, Beaumont, Tex.) of means such as the Student t-test. Owing to inherent varia-
CoaguChek XS PT test system and CoaguChek XS Plus PT tion, laboratory scientists advise that each point-of-care ana-
test system (Roche Diagnostics, Indianapolis, Ind.) lyzers test results be compared with its own reference interval
Gem PCL Plus (Instrumentation Laboratory, Bedford, Mass.) and target therapeutic range.
770 PART VIII Hemostasis and Thrombosis

The pharmacists and nurse practitioners who typically Heparin supports the thrombin-antithrombin reaction
manage anticoagulation clinics appreciate point-of-care instru- through a bridging mechanism (Figure 46-3). If the heparin
ments for their instantaneous turnaround. Patient waiting time molecule exceeds 17 linear saccharide units, thrombin assem-
is eliminated, and dosage adjustment can be made at the time bles on the heparin molecule near the activated antithrombin.
of the patients visit. Clinics so equipped capitalize on the Bridging drives the thrombin-antithrombin reaction at a rate
convenience factor to attract patients and maximize the time four times that of the factor Xa-antithrombin reaction, because
their anticoagulation is within the therapeutic range. factor Xa becomes inactivated only by antithrombins steric
Portable and hand-held capillary specimen point-of-care modification and is not enhanced by bridging. The PTT
devices are designed for patient self-testing. Although self- becomes prolonged within minutes of UFH administration,
testing seldom duplicates central laboratory precision, properly which reflects the immediate anticoagulation effect of UFH.
instructed patients are able to improve the percentage of time UFH preparations vary in average molecular weight, mole-
anticoagulation is within the therapeutic range through fre- cule length, and efficacy. Individual patient metabolic rates
quent self-testing. In the United States, patients are cautioned diverge markedly, because numerous plasma and cellular pro-
not to self-dose, but rather first to discuss dosage adjustments teins bind heparin at varying rates. Consequently, laboratory
with their physicians.14 monitoring is essential.15

Unfractionated Heparin Therapy


UNFRACTIONATED HEPARIN THERAPY AND
Physicians administer UFH intravenously to treat DVT and PE,
THE PARTIAL THROMBOPLASTIN TIME
and to provide initial treatment of AMI; to prevent reocclusion
Heparin Is a Catalyst That Activates after stent placement; and to maintain vascular patency during
Antithrombin to Neutralize Serine Proteases cardiopulmonary bypass surgery with extracorporeal circula-
Standard UFH is a mixture of sulfated glycosaminoglycans tion. Therapy begins with a bolus of 5000 to 10,000 units,
(polysaccharides) extracted from porcine mucosa. The molecu- followed by continuous infusion at approximately 1300 units/
lar weight of UFH ranges from 3000 to 30,000 D. Approxi- hour, adjusted to patient weight. UFH is stopped after 5 days
mately one third of its molecules make available a high-affinity to avoid HIT with thrombosis, a morbid, sometimes fatal side
pentasaccharide that binds plasma antithrombin. The anti effect (see Chapter 42). Most UFH therapy is completed at the
coagulant action of UFH is indirect and catalytic. The time of discharge, typically the second or third day after AMI.
pentasaccharide-bound antithrombin undergoes a steric
change (allostery) which exposes an anticoagulant site that Monitoring Unfractionated Heparin Therapy
covalently binds and inactivates the coagulation pathway Using the Partial Thromboplastin Time
serine proteases factors IIa (thrombin), IXa, Xa, XIa, and XIIa Because of its inherent pharmacologic variations and narrow
(see Chapter 40). Laboratory scientists call activated anti- therapeutic range, UFH therapy is diligently monitored using
thrombin a serine protease inhibitor (serpin), and the protease- the PTT. Blood is collected and assayed before therapy is begun
binding reaction yields the measurable inactive plasma complex to ensure that the baseline PTT is normal.16 A prolonged base-
thrombin-antithrombin. line PTT may indicate a preexisting condition such as the

Fibrin-bound
thrombin

lla

Protease Active
binding protease
AT site site lla

Heparin binding
Heparin binding
AT lla site
site

UFH TAT

AT lla

Figure 46-3 The heparin binding site of antithrombin (AT) binds a specific pentasaccharide, producing an allosteric change that activates AT. Factor IIa
(thrombin) assembles on the heparin surface if the molecule is at least 17 saccharide units long. AT binds IIa and the complex is released from the unfraction-
ated heparin (UFH) to form the soluble, measurable thrombin-antithrombin (TAT) complex. The UFH recycles. Fibrin-bound IIa does not enter the reaction.
CHAPTER 46 Monitoring Antithrombotic Therapies 771

presence of a lupus anticoagulant or a factor deficiency and therapy. The medical laboratory scientist collects 50 or more
confuses the therapeutic interpretation. In such cases, the labo- plasma specimens from patients taking UFH at all levels of
ratory scientist switches to the chromogenic antifactor Xa anticoagulation and measures PTT for all. The specimens must
heparin assay. be from patients who are not receiving simultaneous warfarin
A second specimen is collected at least 4 to 6 hours but not therapy; their PT results must be normal. Chromogenic anti
longer than 24 hours after the bolus dosage that initiates factor Xa heparin assays are performed on all specimens, and
therapy and PTT is measured on this specimen. The result for the paired results are displayed on a linear graph (Figure 46-4).
this specimen should fall within the therapeutic range, which The range in seconds of PTT results that corresponds exactly to
is established by the laboratory scientist (see the next section) 0.3 to 0.7 chromogenic antifactor Xa heparin units/mL is the
and reported with the result. The physician adjusts the infusion therapeutic range.17 This is known as the ex vivo or Brill-Edwards
rate to ensure that the PTT result is within the target range. PTT method for establishing the heparin therapeutic range of the PTT,
measurement is subsequently repeated every 24 hours, and the and its use is required by accrediting agencies. Other approaches
dosage is continually readjusted until UFH anticoagulation is to the determination of the PTT therapeutic range for UFH
complete. The physician also monitors the platelet count daily. therapy are discouraged. For instance, many experts once recom-
A 40% or greater reduction in the count, even within the mended that the therapeutic PTT value be taken as 1.5 to 2.5
normal range, is evidence for HIT (see the discussion of the times the mean of the reference interval. This approach, however,
4Ts HIT diagnosis system in Chapter 42). If HIT is suspected, consistently results in underanticoagulation, which raises the
UFH therapy is immediately discontinued and replaced with risk of a secondary thrombotic event, and must be avoided.
DTI therapy.
Clinical Utility of Partial Thromboplastin
Determining the Partial Thromboplastin Time Monitoring of Unfractionated Heparin
Time Therapeutic Range for Unfractionated Therapy and Reporting of Results
Heparin Therapy The medical laboratory scientist reports the PTT results, the
The hemostasis laboratory is required to establish and com- reference interval, and the UFH therapeutic range to the clini-
municate a therapeutic range for PTT for management of UFH cian (physician, nurse, or pharmacist) who is managing the

HEPARIN THERAPEUTIC RANGE


180
170
160
150
140
130
120
110
PTT

100
90
80
70

60
50
40
30
20
10
0
0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0
Anti-Xa
Figure 46-4 Laboratory scientists establish the partial thromboplastin time (PTT) therapeutic range for unfractionated heparin (UFH) by collecting specimens
from at least 50 patients receiving UFH at representative dosages who have normal PTs and at least 20 individuals not receiving heparin. PTT and chromogenic
antifactor Xa heparin assays are performed on all specimens, and a linear graph of paired results is prepared with PTT on the vertical scale. The PTT range
in seconds is correlated with the chromogenic antifactor Xa therapeutic range of 0.3 to 0.7 units/mL or the prophylactic range of 0.1 to 0.4 units/mL.
772 PART VIII Hemostasis and Thrombosis

patients UFH dosage. Because reagent sensitivity varies among administered to yield results of 180 to 240 seconds in DVT or
producers and among individual producers reagent lots, the 400 seconds during cardiopulmonary bypass surgery or percu-
clinician must evaluate PTT results in relation to the institu- taneous cardiac intervention.
tions therapeutic and reference intervals.18 No mean analogous The following instruments are designed to provide semiau-
to the INR exists for normalizing PTT results, because reagents tomated ACT results and are popular in operating suites:
are too variable.19 The PTT is used most often to measure the Cascade POC, Actalyke XL and Actalyke Mini II (Helena
effects of UFH therapy. LMWH selectively catalyzes the neutral- Point of Care, Beaumont, Tex.)
ization of factor Xa over that of thrombin, and its effects cannot Gem PCL Plus (Instrumentation Laboratory, Bedford,
be measured using the PTT. The chromogenic antifactor Mass.)
Xa heparin assay may be used to assay UFH, LMWH, and HMS Plus, ACT Plus (bench top, Medtronic Cardiac Surgery,
fondaparinux.20 Minneapolis, Minn.)
INRatio2 PT INR monitor (HemoSense/Inverness Medical,
Limitations of Partial Thromboplastin San Diego, Calif.)
Time Monitoring of Unfractionated ProTime Microagulation SystemNew ProTime; Hemo-
Heparin Therapy chron Signature Elite; Hemochron Signature Plus; Hemo-
Several conditions render the patient unresponsive to heparin chron Response (International Technidyne Corporation,
therapy, a circumstance called heparin resistance.21 Inflamma- Edison, N.J.)
tion typically is accompanied by hyperfibrinogenemia, with a For the Hemochron equipment, after specimen collection
fibrinogen level of greater than 498mg/dL, and coagulation the tube is placed in the instrument, where it is rotated and
factor VIII activity of greater than 186% of normal. Both tend continuously monitored. When a clot forms, a magnet posi-
to shorten the PTT and render it less sensitive to the effects of tioned within the sample is pulled away from a sensing device,
heparin. In many patients, the antithrombin level becomes which stops the timer. The time interval to clot formation is
depleted as a result of prolonged therapy or an underlying recorded automatically. The results of the ACT assay are com-
deficiency secondary to chronic inflammation. In this instance, parable to those of the PTT assay for UFH monitoring, pro-
the PTT result remains within the reference interval or becomes vided adequate quality control steps are taken. The ACT is
only slightly prolonged despite ever-increasing heparin dosages. particularly useful for monitoring the effects of the high blood
Inflammation may be reduced through administration of levels (1 to 2 units/mL) of heparin during coronary artery
steroids and aspirin or nonsteroidal antiinflammatory drugs, bypass surgery.
and antithrombin concentrate may be given. In the interim,
however, use of an alternative monitoring test such as the Reversal of Unfractionated Heparin Overdose
chromogenic antifactor Xa heparin assay is necessary. Using Protamine Sulfate
Platelets in anticoagulated whole-blood specimens release Protamine sulfate, a cationic protein extracted from salmon
platelet factor 4, a heparin-neutralizing protein (see Chapter sperm, neutralizes UFH at a ratio of 100 units of heparin per
13). In specimens from patients receiving heparin therapy, the milligram of protamine sulfate. The physician administers
PTT begins to shorten as soon as 1 hour after collection because protamine sulfate slowly by intravenous push. The effect of the
of in vitro platelet factor 4 release unless the specimen is cen- protamine sulfate may be detected by the shortening of the PTT
trifuged and the platelet-poor plasma is removed from the cells or ACT to within the reference interval. Protamine sulfate also
(see Chapter 45).22 Hypofibrinogenemia, factor deficiencies, neutralizes LMWH, although the neutralization is incompletely
and the presence of lupus anticoagulant, fibrin degradation reflected in the results of the chromogenic antifactor Xa
products, or paraproteins prolong the PTT independent of heparin assay described in the following paragraphs.
heparin levels.23

LOW-MOLECULAR-WEIGHT HEPARIN
Monitoring Unfractionated Heparin Therapy
THERAPY AND THE CHROMOGENIC
Using the Activated Clotting Time
ANTIFACTOR XA HEPARIN ASSAY
The ACT is a 1966 modification of the Lee-White whole-blood
clotting time test that is used to monitor high-dose heparin Low-Molecular-Weight Heparin Is Derived
therapy (see Chapter 45). The ACT is measured at the clinic, at from Unfractionated Heparin
the inpatients bedside, in the cardiac catheterization labora- Uncertainty about UFH dose response and the ever-present
tory, or in the surgical suite.24 threat of HIT led to the development of LMWH, which was
ACT assay distributors provide evacuated blood specimen approved for anticoagulant prophylaxis in the United States
collection tubes that contain 12mg of diatomaceous earth, a and Canada in 1993. LMWH is prepared from UFH using
particulate clot activator. The negative pressure within the tube chemical (enoxaparin) or enzymatic (tinzaparin) fraction-
is calibrated to collect 2mL of blood. As soon as the specimen ation. Fractionation yields a product with a mean molecular
is collected, the phlebotomist starts a timer, and the tube is weight of 4500 to 5000 D, about one third the mass of UFH.
inverted a few times to disperse the activator. The phleboto- LMWH possesses the same active pentasaccharide sequence
mist continues to invert the tube until a clot forms. The median as UFH; however, the overall shorter polysaccharide chains
of the ACT reference interval is 98 seconds. Heparin is provide little space for thrombin bridging, so the thrombin
CHAPTER 46 Monitoring Antithrombotic Therapies 773

Platelet

Xa
Protease Active Platelet
binding protease
AT site site Xa

Heparin binding
site No heparin
AT Xa binding site

LMWH

AT Xa

Figure 46-5 The antithrombin (AT) binding site binds a specific heparin pentasaccharide, producing an allosteric change that activates AT. The low-
molecular-weight heparin (LMWH) molecule is too short to support a factor IIaAT reaction; however, the activated AT binds factor Xa independently of the
bridging phenomenon, producing a soluble ATfactor Xa complex. The heparin recycles. Platelet-bound Xa does not enter the reaction.

neutralization response is reduced (Figure 46-5). The anti- accumulation in plasma. LMWH therapy in children, morbidly
thrombin to factor Xa neutralization response is unchanged, obese adults, and underweight adults as well as therapy during
however, because the Xa reaction does not rely on bridging, so pregnancy also require monitoring because of fluid compart-
LMWH provides nearly the same anticoagulant efficacy as UFH, ment imbalances.
although predominantly through Xa inhibition. A specimen is collected 4 hours after subcutaneous injec-
LMWH is administered by subcutaneous injection once or tion, and the plasma is tested using the chromogenic anti
twice a day using premeasured syringes at selected dosages, factor Xa heparin assay, because the PTT is insensitive to LMWH
for instance, 30mg subcutaneously every 12 hours or 40mg effects. The chromogenic antifactor Xa heparin assay employs
subcutaneously once daily. Prophylactic applications provide a reagent that provides a fixed concentration of factor Xa and
coverage during or after general and orthopedic surgery and substrate specific to factor Xa (Figure 46-6). Some distributors
trauma, typically for 14 days from the time of the event. LMWH add fixed concentrations of antithrombin; others rely on the
also is used to treat DVT, PE, and unstable angina. LMWH is patients plasma antithrombin. The latter formulation provides
indicated during pregnancy for women at risk of VTE, because sensitivity to antithrombin depletion or deficiency. Heparin
warfarin, which causes birth defects, cannot be used. When forms a complex with reagent Xa and antithrombin; a mea-
patients who are taking warfarin require surgery, warfarin sured excess of factor Xa digests the substrate, which yields a
is discontinued for up to a week before the procedure and colored product whose intensity is inversely proportional to
replaced with LMWH.25 heparin concentration.
The advantages of LMWH are rapid bioavailability after sub- To prepare a standard curve, the laboratory scientist obtains
cutaneous injection, which makes intravenous administration characteristic heparin from the pharmacy and computes and
unnecessary; a half-life of 3 to 5 hours compared with 1 to 2 prepares dilutions that correspond with the therapeutic range.
hours for UFH; and a fixed dose response that reduces the need If the chromogenic antifactor Xa heparin assay is to be used
for laboratory monitoring. The risk of HIT is reduced by 90% to monitor UFH and LMWH, a single hybrid standard curve
in people who have never received heparin before; however, may be prepared.26,27 A separate curve may be necessary to
LMWH may cross-react with previously formed antibodies monitor pentasaccharide. The therapeutic range is 0.5 to 1.0
against heparinplatelet factor 4. Consequently, LMWH is con- unit/mL for twice-daily regimens and 1 to 2 units/mL for once-
traindicated in patients who previously developed HIT after daily regimens.
UFH therapy. The risk of LMWH-induced bleeding is equiva- The chromogenic antifactor Xa heparin assay is the only
lent to that for UFH, about 10%. assay available to monitor LMWH and pentasaccharide therapy.
It also may be used in place of the PTT with little or no modi-
Monitoring Low-Molecular-Weight fication to assay UFH and substitutes for the PTT when clinical
Heparin Therapy or laboratory conditions render PTT results unreliable. The
LMWH is cleared by the kidneys alone, so it accumulates in chromogenic antifactor Xa heparin assay is tertiary in the
renal insufficiency. Laboratory monitoring of LMWH therapy sense that it measures the concentration of heparin and not its
is necessary when the creatinine clearance is less than 30mL/ anticoagulant effects; however, the assay is precise and, in con-
min or serum creatinine is greater than 4mg/dL. During trast to the PTT, is affected by few interferences. The chromo-
LMWH therapy, creatinine assays are performed periodically genic antifactor Xa heparin assay is also the reference method
to document kidney function and avoid the risk of LMWH for establishing the PTT therapeutic range. Laboratory directors
774 PART VIII Hemostasis and Thrombosis

Heparin

AT + AT

Xa
AT + Xa AT

+ Xa + Substrate Substrate

Figure 46-6 The chromogenic antifactor Xa heparin assay. The reagent is a mixture of antithrombin (AT) and a measured excess of Xa. (Some kits do
not provide AT and rely solely on patient plasma AT.) AT binds heparin, and this complex binds factor Xa. Excess free factor Xa digests its substrate to produce
a colored end product. The color intensity of the product is inversely proportional to plasma heparin. This assay is used for unfractionated heparin, low-
molecular-weight heparin, and pentasaccharide (fondaparinux); a dedicated standard curve is required for fondaparinux.

D-glucosamine D-iduronic acid

OSO2Na COONa OSO2Na OSO2Na

O O O O O
COONa
OH OH O OSO2Na O
OH OH
O O
HO OMe

NHSO2Na NHSO2Na OSO2Na NHSO2Na


OH

D-glucuronic acid
Figure 46-7 The specific saccharide sequence in unfractionated heparin and low-molecular-weight heparin (glucosamine, glucuronic acid, glucosamine,
iduronic acid, glucosamine) is synthesized to make fondaparinux, which binds and activates the heparin-binding site of antithrombin. (From Turpie AGG:
Pentasaccharides, Semin Hematol 39:159-171, 2002.)

have begun to recognize the merits of the chromogenic anti contraindicated in patients with creatinine clearance of less
factor Xa heparin assay and substitute it for the PTT in monitor- than 30mL/min and has not been cleared for patients with
ing UFH therapy as well as in monitoring therapy with LMWH body weights of less than 110lb.28
and fondaparinux. All that is lacking to support a complete The chromogenic antifactor Xa heparin assay is used to
switchover to the chromogenic antifactor Xa heparin assay is monitor off-label fondaparinux therapy in infants, children,
a volume-based reduction in assay cost and enactment of obese or underweight patients, patients receiving treatment
reasonable reimbursement policies. for more than 7 to 8 days, and pregnant women. Blood is col-
lected 4 hours after injection, and the target range is 0.14 to
0.19mg/L. The laboratory scientist prepares a standard curve
MONITORING PENTASACCHARIDE THERAPY
using fondaparinuxnot UFH, LMWH, or a hybrid standard
USING THE CHROMOGENIC ANTIFACTOR
because concentrations are expressed in milligrams per millili-
XA HEPARIN ASSAY
ter and not units per deciliter. The PTT is not sensitive to the
Fondaparinux sodium (Arixtra; GlaxoSmithKline, Research effects of fondaparinux because, although fondaparinux reacts
Triangle Park, N.C.) is a synthetic formulation of the active with antithrombin and factor Xa, it does not inhibit thrombin,
pentasaccharide sequence in UFH and LMWH (Figure 46-7). IXa, XIa, or XIIa.
Fondaparinux raises antithrombin activity 400-fold. It is equiv-
alent in clinical efficacy and safety to UFH and LMWH and
RIVAROXABAN, AN ORAL DIRECT
has a reproducible dose response and a half-life of 12 to 17
FACTOR Xa INHIBITOR
hours. It is administered as once-a-day subcutaneous injections
of 2.5mg. Fondaparinux is approved for prophylaxis after Oral rivaroxaban (Xarelto; Bayer Healthcare AG, Leverkusen,
surgery and for the treatment of DVT and PE, but its use is Germany; Ortho-McNeil Pharmaceuticals, Inc., Raritan, N.J.) is
CHAPTER 46 Monitoring Antithrombotic Therapies 775

an oxazolidinone derivative that directly inhibits factor Xa thrombin (argatroban and bivalirudin) without involving anti-
independently of antithrombin.24 It functions unmodified after thrombin (Figure 46-8). DTIs are substituted for UFH or
ingestion, as it inhibits free Xa, Xa bound by IXa, and clot- LMWH when HIT is suspected or confirmed using the 4Ts
bound Xa. Rivaroxaban has favorable efficacy and safety out- assessment system (see Chapter 42). Without the use of a DTI,
comes compared with LMWH or warfarin. Rivaroxaban was the risk of thrombosis is 50% for the next 30 days after heparin
cleared in September 2008 by both Health Canada and the withdrawal.
European Commission for VTE prophylaxis for use in patients Argatroban (Novostan; GlaxoSmithKline, Research Triangle
who are undergoing total knee replacement or total hip replace- Park, N.C.) is a non-protein L-arginine derivative with a molec-
ment surgery.29 The standard oral dosage is 10mg/day, and ular weight of 527 D. Argatroban was approved in 1997 for
because of its predictable pharmacodynamics, laboratory mon- anticoagulation in HIT, in which warfarin, UFH, and LMWH
itoring is seldom required. As of November 2010, rivaroxaban are contraindicated.30 Argatroban is approved for prophylaxis,
awaited clearance by the U.S. FDA, although approval was for treatment, and also for anticoagulation during cardiac
recommended by an FDA study committee in March 2009. catheterization for patients with HIT.
Rivaroxaban prolongs the PT and PTT, as reported in several The physician initiates the argatroban intravenous infusion
clinical trials; however, no attempt has been made to correlate at 2mcg/kg/min or in patients with hepatic disease at
PT and PTT results with clinical efficacy. Rivaroxaban may also 0.5mcg/kg/min. During percutaneous cardiac intervention, a
be assayed using the chromogenic antifactor Xa heparin assay bolus of 350mcg/kg is given over 3 to 5 minutes followed by
by standardizing with rivaroxaban in place of UFH, LMWH, or 25mcg/kg/min. Argatroban is cleared by the liver and excreted
fondaparinux, although it will be necessary for laboratory in stool. There is a 5% general bleeding risk and no direct
scientists to correlate laboratory results with clinical outcomes. antidote; however, the half-life is 51 minutes, and argatroban
Rivaroxaban appears to be an attractive alternative to the clears from the blood in 2 to 4 hours. Argatroban therapy
ever-dangerous warfarin and inconvenient LMWH injections requires laboratory monitoring, particularly for detection of an
because it is safer, is more effective, is convenient, and requires overdose.
little laboratory monitoring. Rivaroxaban may be the first of
several oral anticoagulants that function by inhibiting either Lepirudin, a Recombinant Analogue of Hirudin
factor Xa or IIa. Another promising anti-Xa anticoagulant in Lepirudin (Refludan; ZLB Behring GmbH, Montville, N.J., and
advanced clinical trials is apixaban. Dabigatran is an oral DTI Marburg, Germany) is a recombinant 7000-D protein DTI.
(anti-IIa anticoagulant) that is described in the next section. Lepirudin is an analogue of natural hirudin, which is produced
in trace amounts by the medicinal leech Hirudo medicinalis.
Lepirudin is approved for anticoagulation in HIT. It binds free
MONITORING DIRECT THROMBIN
but not fibrin-bound thrombin, because the drug molecule is
INHIBITOR THERAPY USING THE PARTIAL
too large and is sterically hindered from binding clot-bound
THROMBOPLASTIN TIME OR ECARIN
thrombin.31
CLOTTING TIME
The physician administers a bolus of 0.4mg/kg for patients
Argatroban up to 110kg over 15 to 20 seconds followed by 0.15mg/kg/hr
The DTIs argatroban, lepirudin, and bivalirudin reversibly bind as a continuous intravenous infusion for 2 to 10 days. Labora-
and inactivate free thrombin (all three agents) and clot-bound tory monitoring is necessary because the risk of bleeding is

Active
protease
site

Argatroban Fibrin
lla

HN 0 CO2H

H2N N
N
NH
H
CH3
H2O O2S
H
N
CH3 lla
lla

Free thrombin Clot-bound


thrombin
Figure 46-8 Argatroban inhibits the active site of free and clot-bound thrombin. Antithrombin is not involved in this reaction.
776 PART VIII Hemostasis and Thrombosis

10%. Lepirudin is cleared by the kidney. The dosage must Monitoring Direct Thrombin Inhibitor Therapy
be reduced in the presence of elevated serum creatinine or DTIs prolong the thrombin time, PT, PTT, and ACT.36 For non-
reduced creatinine clearance and is discontinued when creati- surgical therapy, distributors of argatroban, lepirudin, and
nine clearance is less than 15mL/min or serum creatinine is bivalirudin recommend monitoring with the PTT using the
greater than 6mg/dL. Serial measurement of creatinine levels target therapeutic range of 1.5 to 3.0 times the mean of the
is necessary throughout lepirudin therapy. There is no antidote laboratory reference interval. Blood is collected 4 hours after
for lepirudin in overdose, but the plasma half-life is only the initiation of intravenous therapy for bivalirudin or lepiru-
20 minutes. din and 2 hours after therapy initiation for argatroban, and the
Antihirudin antibodies form in about 40% of HIT patients dosage is adjusted to achieve a PTT in the therapeutic range.
treated with lepirudin. These antibodies may increase the anti- The ACT assay is used during cardiac catheterization or coro-
coagulant effect, because they actually delay the renal elimina- nary artery bypass graft surgery. During these procedures, the
tion of active lepirudin-antihirudin complexes while the target ACT for bivalirudin therapy is 320 to 400 seconds
anticoagulant remains active. Strict laboratory monitoring is (median normal value, 98 seconds). There are neither recom-
necessary during prolonged therapy, and anaphylaxis may mendations nor therapeutic target ranges for monitoring of
occur during a second administration.32 dabigatran therapy.
In instances in which the baseline PTT is prolonged by
Bivalirudin, a Recombinant Derivative lupus anticoagulant or factor deficiencies, the Ecarin clotting
of Hirudin time (ECT) is an attractive alternative for monitoring DTI
Bivalirudin (Angiomax; The Medicines Company, Parsippany, therapy, including dabigatran therapy. Ecarin (ecarinase;
N.J.) is a synthetic 20-amino acid peptide derivative of hirudin Pentapharm, Basel, Switzerland) is an enzyme extracted from
with a molecular weight of 2180 D. Bivalirudin inactivates both Echis carinatus venom that converts prothrombin to the inter-
free and clot-bound thrombin. Bivalirudin was cleared in 2000 mediate molecule meizothrombin, which converts fibrinogen
for use as an anticoagulant in patients with unstable angina to fibrin. DTIs bind meizothrombin and generate a linear,
at risk for HIT who are undergoing percutaneous coronary dose-dependent prolongation of the ECT. Aside from DTI
intervention.33 administration, the ECT is prolonged only by abnormally low
Bivalirudin is intended for use with concurrent aspirin prothrombin and fibrinogen activity. A more recently described
therapy at a dosage of 325mg/day and has been studied only ECT reagent enriched with prothrombin and fibrinogen elimi-
in patients receiving aspirin.34 Physicians provide an intrave- nates prolongation caused by variations in patient prothrom-
nous bolus dose of 0.75mg/kg followed by an infusion of bin and fibrinogen levels to permit accurate determinations of
1.75mg/kg/hr for the duration of the percutaneous cardiac DTI effects.37
intervention. After 4 hours, an additional intravenous infusion Ecarin reagent requires special handling, and the assay has
may be given at a rate of 0.2mg/kg/hr for 20 hours. no prepared calibrators or controls, so the ECT test is only
The rate of major hemorrhage with bivalirudin is 4%. There slowly becoming available in acute care facility laboratories.
is no antidote; however, in patients with normal renal function Specialized references laboratories such as Esoterix Coagula-
the half-life is 25 minutes. The dosage is decreased in patients tion in Aurora, Colorado, offer the assay.
with reduced creatinine clearance or elevated serum creatinine.
No antihirudin-type antibody has been detected during
MONITORING ANTIPLATELET THERAPY
bivalirudin therapy.
USING PLATELET ACTIVITY ASSAYS
Dabigatran, an Oral Direct Intravenous Glycoprotein IIb/IIIa Inhibitors
Thrombin Inhibitor Are Used During Cardiac Catheterization
Dabigatran etexilate (Pradaxa; Boehringer Ingelheim, Ingel- Glycoprotein IIb and glycoprotein IIIa are present on the mem-
heim, Germany) is an oral prodrug that converts upon diges- brane of resting platelets. Upon activation by any agonist, these
tion to active dabigatran, a reversible DTI that binds both free molecules join to form glycoprotein IIb/IIIa complexes, recep-
and clot-bound thrombin. Dabigatrans efficacy and safety tors that bind fibrinogen and von Willebrand factor through
appear to match those of LMWH and warfarin, and it has no their arginineglycineaspartic acid (RGD) sequences. Fibrino-
interaction with food. It is cleared by the kidneys, has a half-life gen binding to glycoprotein IIb/IIIa supports the key step of in
of 12 to 17 hours, and is not metabolized by liver cytochrome vivo platelet aggregation (see Chapter 13). The intravenous
enzymes.35 Dabigatran was cleared for use in Europe and glycoprotein IIb/IIIa inhibitors (GPIs) abciximab, eptifibatide,
Canada in the spring of 2008 for VTE prophylaxis during total and tirofiban fill glycoprotein IIb/IIIa receptor sites and
knee replacement or total hip replacement surgery at dosages block fibrinogen or von Willebrand factor binding, thereby
of 220mg/day, or 150mg/day in the elderly or those with effectively preventing platelet aggregation. The GPIs are used
moderate renal impairment. In October 2010, Dabigatran was to maintain vascular patency during cardiac catheterization
cleared for prophylaxis in atrial fibrillation by the U.S. FDA. and stent placement.
The question of laboratory monitoring in pregnant patients, Abciximab (ReoPro; Eli Lilly and Company, Indianapolis,
the elderly, morbidly obese or underweight adults, and patients Ind.) is the 47,615-D Fab fragment of a mouse monoclonal
with renal dysfunction has not been addressed. antibody specific for glycoprotein IIb/IIIa that effectively fills
CHAPTER 46 Monitoring Antithrombotic Therapies 777

the receptor site.38 The dose is 0.25mg/kg given by intravenous minutes and then continued at 0.1mcg/kg/min throughout
bolus administered 10 to 60 minutes before the start of cardiac catheterization and for 12 to 24 hours after catheteriza-
cardiac catheterization, followed by continuous infusion of tion. Tirofiban is excreted through the kidney, so the dosage is
0.125mcg/kg/min for up to 12 hours. Abciximab is always halved when the creatinine clearance is less than 30mL/min.
coadministered with UFH and aspirin. Before infusion the PT, Both eptifibatide and tirofiban are monitored using the same
PTT, ACT, hemoglobin, hematocrit, and platelet count are laboratory protocol as that for abciximab, and both are discon-
determined to detect any hemostatic or hematologic abnor- tinued if the platelet count drops by 25% or more.
mality.39 During infusion, the PTT and ACT are maintained
within the UFH therapeutic range as determined by the labora- Aspirin, Clopidogrel, Ticlopidine, and
tory. Platelet counts are performed at 2 hours, 4 hours, and 24 Prasugrel Reduce the Incidence of
hours following the initial bolus. If the platelet count drops by Arterial Thrombosis
25% or more, UFH and abciximab are discontinued and the The most commonly prescribed oral antiplatelet drugs are
platelet count is monitored daily until it returns to within the aspirin, clopidogrel (Plavix; Sanofi-Aventis, Bridgewater, N.J.,
reference interval. Abciximab and aspirin efficacy may each be and Bristol-Myers Squibb, New York, N.Y.), ticlopidine (Ticlid;
measured using the Multiplate analyzer or VerifyNow IIb/IIIa Roche Pharmaceuticals, Nutley, N.J.), and prasugrel (Effient; Eli
assay. Aspirin efficacy may be determined using the PFA-100 or Lilly and Company, Indianapolis, Ind.). Aspirin irreversibly
AspirinWorks assay. acetylates the platelet enzyme cyclooxygenase at the serine at
Eptifibatide (Integrilin; Schering Corporation, Kenilworth, position 529 (Figure 46-9). The serine-bound acetyl group ste-
N.J.), is an 832-D heptapeptide that binds and blocks access rically hinders the access of arachidonic acid to its reactive site
to platelet glycoprotein IIb/IIIa.40 It is coadministered with within the cyclooxygenase molecule. This prevents production
aspirin and UFH. An intravenous bolus of 180mcg/kg is given of platelet-activating thromboxane A2 through the eicosanoid
as soon as possible after initial diagnosis, and a continuous synthesis pathway (see Chapter 13).42 Acetylation is irrevers-
intravenous drip of 2mcg/kg/min is continued for up to 96 ible; the eicosanoid synthesis pathway is shut down for the
hours following the initial bolus, including throughout cardiac remainder of the life of the platelet.
catheterization. In contrast, ticlopidine, clopidogrel, and prasugrel, which
Tirofiban hydrochloride (Aggrastat; Baxter Healthcare Cor- are members of the thienopyridine drug family, reversibly
poration, Deerfield, Ill.), is a 495-D nonpeptide GPI that is occupy the platelet membrane adenosine diphosphate (ADP)
coadministered with aspirin and UFH.41 It is administered receptor P2Y12, suppressing the normal platelet aggregation
intravenously at an initial rate of 0.4mcg/kg/min for 30 and secretion response to the activating ligand (agonist), ADP.

Ticlopidine, clopidogrel,
and prasugrel reversibly
bind P2Y12
Ph
osp
ho
lipa
se
A
Aspirin irreversibly 2 P2Y 12
acetylates COX 1

Arachidonic acid
COOH
Cyclooxygenase-1

PGG2/PGH2

TXA Synthase

Thromboxane A2
a
/III
IIb
GP

Thromboxane B2 is a stable,
Abciximab, eptifibatide, measurable plasma product that
and tirofiban irreversibly is reduced in aspirin therapy
bind GP IIb/IIIa

Figure 46-9 Antiplatelet drugs employ three mechanisms to inactivate platelets. Aspirin irreversibly acetylates and inactivates cyclooxygenase 1 (COX-1).
Ticlopidine, clopidogrel, and prasugrel reversibly bind the adenosine diphosphate (ADP) receptor, P2Y12. Intravenous abciximab, eptifibatide, and tirofiban
irreversibly bind the fibrinogen binding site, glycoprotein (GP) IIb/IIIa. PGG2, Prostaglandin G2; PGH2, prostaglandin H2; TXA synthase, thromboxane A2
synthase.
778 PART VIII Hemostasis and Thrombosis

Aspirin is often prescribed alone at 81 or 325mg/day to treatment failure and the need to revise dosage or change
prevent myocardial infarction and ischemic cerebrovascular to a new antiplatelet drug. All three Accumetrics VerifyNow
disease in patients with stable or unstable angina,43,44 AMI,45 systems have been FDA cleared.
transient cerebral ischemia,46 peripheral vascular disease,47 or Multiplate (DiaPharma Group, Inc., West Chester, Ohio):
stroke.48,49 In healthy people, aspirin prophylaxis annually pre- The Multiplate analyzer is automated for point-of-care
vents four thrombotic events per 1000 individuals treated, testing and uses impedance aggregometry to simultaneously
although it carries a risk of bleeding.50-52 or individually test for platelet aggregation responses to
Ticlopidine has been prescribed at 250mg twice a day con- aspirin, clopidogrel, and GPIs.62 The Multiplate requires
currently with aspirin, but has been largely replaced by clopi- 300mcL of whole blood. The aspirin resistance assay uses
dogrel and prasugrel. Ticlopidine has been associated with a arachidonic acid as its agonist, the clopidogrel response
risk of thrombotic thrombocytopenic purpura.53 assay uses ADP, and the GPI response uses TRAP. The instru-
Clopidogrel is prescribed at 75mg/day together with aspirin ment integrates three aggregometry parameters, aggregation
at 81 or 325mg/day. Patients appear to metabolize clopidogrel velocity, maximum aggregation, and area under the aggregation
at varying rates, which raises the need for routine laboratory curve to produce arbitrary units. The local laboratory estab-
monitoring using platelet function assays. lishes reference interval limits and expected therapeutic
Prasugrel was cleared by the FDA in July 2009.54,55 It is target levels. Results that are outside the therapeutic target
administered as an oral prodrug that is converted in the liver range indicate possible treatment failure and the need to
via several cytochrome P-450 pathways to an active metabolite revise dosage or change to a new antiplatelet drug. The Multi
whose elimination half-life is 2 to 15 hours. Treatment begins plate is available in Europe; the DiaPharma Group is cur-
with a single 60-mg oral loading dose and continues at 10mg rently awaiting United States FDA clearance for the device.
daily, or 5mg daily for patients who weigh less than 60kg. PFA-100 (Siemens Healthcare Diagnostics, Inc., Deerfield,
Prasugrel is prescribed to be taken with aspirin at 81mg or Ill.): The PFA-100 system uses two cartridges: the first pro-
325mg daily and appears to require no laboratory monitoring. vides an aperture impregnated with collagen and epineph-
Prasugrel carries a higher risk of bleeding than clopidogrel and rine, and the second provides an aperture impregnated with
may be associated with an increased risk of solid tumors. collagen and ADP. The PFA-100 requires 800mcL whole
blood per cartridge. The specimen passes through the aper-
Variable Antiplatelet Response and ture until activation by the agonist causes closure, a param-
Laboratory Monitoring of eter called closure time. The PFA-100 tests only for aspirin
Antiplatelet Resistance resistance. A closure time that is shorter than the anticipated
Several investigations confirm that 10% to 20% of people who therapeutic range for aspirin indicates resistance.
are taking aspirin generate an inadequate response as measured AspirinWorks (Corgenix, Inc., Denver, Colo.): The Aspirin-
in the laboratory by light transmittance platelet aggregometry Works immunoassay measures a urine metabolite of platelet
or whole-blood lumiaggregometry using arachidonic acid as eicosanoid synthesis and thromboxane A2 activation (see
the agonist. Inadequate response to aspirin has been termed Chapter 13). Hepatocyte 11-hydroxythromboxane dehydroge-
aspirin resistance.56-60 Likewise, the response to clopidogrel as nase acts upon stable platelet-derived plasma thromboxane
measured by aggregometry using ADP as the agonist varies B2, the end product of eicosanoid synthesis and the stable
markedly among patients, and the results of this assay may be analogue of thromboxane A2, to produce water-soluble
used to adjust clopidogrel dosage. Mechanisms that explain 11-dehydrothromboxane B2.63 The urine concentration of
aspirin resistance and clopidogrel response variation are cur- 11-dehydrothromboxane B2 is high and, because platelets
rently under study. Unlike treatment with aspirin and clopido- seem to be its primary source, proportionally reflects platelet
grel, prasugrel therapy may not require laboratory monitoring. activity within the previous 12 hours. Urine levels of
Aggregometry is the reference method for determining aspirin 11-dehydrothromboxane B2 frequently are elevated in
and clopidogrel responses, but more rapid assays are available, atherosclerosis; after stroke, transient ischemic attack, or
as follows: intracerebral hemorrhage; and in atrial fibrillation. Levels
VerifyNow (Accumetrics, San Diego, Calif.): The Accumetrics of 11-dehydrothromboxane B2 typically are decreased in
VerifyNow system is designed for point-of-care testing and patients receiving aspirin therapy, even in those with athero-
uses light transmittance aggregometry to individually test sclerosis, myocardial infarction, and atrial fibrillation, but
for platelet aggregation responses to aspirin, clopidogrel, appear to remain normal in patients with aspirin resistance.
and GPIs.61 For each assay a cartridge is provided that con-
tains the desired agonist and fibrinogen-coated beads. Veri-
FUTURE OF ANTITHROMBOTIC THERAPY
fyNow Aspirin uses arachidonic acid as its agonist. VerifyNow
P2Y12 uses ADP. VerifyNow IIb/IIIa uses thrombin receptor Antithrombotic therapy, unchanged for 50 years, is likely to see
activating polypeptide (TRAP), which activates platelets by major changes between 2011 and 2020. As described earlier,
binding to the thrombin receptor protease-activated receptor several oral anticoagulants currently under development or
1. The laboratory establishes reference interval limits and awaiting clearance are likely to replace warfarin, the heparins,
therapeutic target limits for each assay. Results that are and fondaparinux. Likewise, a series of new and emerging
outside the therapeutic target range indicate possible antiplatelet drugs will augment the time-honored aspirin tablet.
CHAPTER 46 Monitoring Antithrombotic Therapies 779

The work of the clinical laboratory will reflect these changes, of. Antiplatelet response monitoring will grow in convenience
moving from the PT and PTT to chromogenic antifactor Xa, and take advantage of flow cytometry, immunoassays, and
chromogenic X, and new molecular assays currently undreamed molecular assays that are currently in development.

SUMMARY
Warfarin is the only oral anticoagulant currently available. It was The DTIs argatroban, lepirudin, and bivalirudin bind thrombin
developed in 1940 by Link and first used in 1952. It prevents VTE, without involving antithrombin and are substituted for heparin in
but it has a narrow therapeutic range, and an overdose causes patients with HIT. Bivalirudin is used only in combination with
hemorrhage. Warfarin therapy is monitored by the PT assay and aspirin. DTI therapy is monitored using the PTT or ECT, and all
reported as an INR. PT measurement is available on portable affect the PT results during switchover to warfarin therapy. Dabi-
point-of-care instrumentation. gatran is the first of several oral DTIs that may soon replace
UFH is administered intravenously to provide immediate control of warfarin, the heparins, and fondaparinux.
coagulation, especially during cardiac catheterization or coronary The antiplatelet drugs aspirin, prasugrel, clopidogrel, and ticlopi-
artery bypass graft surgery involving extracorporeal circulation. Its dine are used after arterial thrombotic events to prevent repeat
therapeutic effect is monitored using PTT testing, which requires AMI, stroke, and peripheral artery occlusion. Patient responses to
the scientist to develop a therapeutic range in seconds keyed to antiplatelet therapy vary. Response variation is detected using
the chromogenic antifactor Xa heparin assay. PTT results are platelet aggregometry, the Accumetrics VerifyNow system, the
subject to several interferences. Multiplate system, or the AspirinWorks assay.
LMWH and pentasaccharide substitute for UFH and are admini The intravenous antiplatelet drugs abciximab, eptifibatide, and
stered subcutaneously for both prophylaxis and therapy. Both tirofiban are used during cardiac catheterization to maintain vas-
provide rapid bioavailability, predictable dose response, and cular patency. Because they may cause thrombocytopenia, the
longer half-lives than UFH. LMWH and pentasaccharide therapy platelet count is monitored carefully. Their efficacy may be moni-
require laboratory monitoring in patients with renal insufficiency, tored using the Accumetrics VerifyNow system or the Multiplate
pregnant women, morbidly obese patients, children, and under- system.
weight adults using the chromogenic antifactor Xa heparin assay.
Rivaroxaban is the first to come to market of several oral direct Now that you have completed this chapter, go back and
antifactor Xa anticoagulants that require little laboratory monitor- read again the case study at the beginning and respond
ing and may soon replace warfarin, the heparins, and fondaparinux. to the questions presented.

R E V I E W Q UESTIONS
1. What is the PT INR therapeutic range for warfarin therapy 3. What is the greatest advantage of point-of-care PT testing?
when a patient has a mechanical heart valve? a. It permits self-dosing of warfarin.
a. 1 to 2 b. It is inexpensive.
b. 2 to 3 c. It is convenient.
c. 2.5 to 3.5 d. It is precise.
d. Warfarin is not indicated for patients with mechanical
heart valves 4. You collect a citrated whole-blood specimen to monitor
UFH therapy. What is the longest it may stand before the
2. Monitoring of a patient taking warfarin showed that her plasma must be separated from the cells?
anticoagulation results remained stable over a period of a. 1 hour
about 7 months. The frequency of her visits to the labora- b. 4 hours
tory began to decrease, so the period between testing aver- c. 24 hours
aged 6 weeks. This new testing interval is: d. Indefinitely
a. Acceptable for a patient with stable anticoagulation
results after 6 months 5. What test is used to monitor high-dose UFH therapy in the
b. Unnecessary because monitoring for patients taking cardiac catheterization operating suite?
oral anticoagulants can be discontinued entirely after a. PT
4 months of stable test results b. PTT
c. Too long even for a patient with previously stable test c. Bleeding time
results; 4 weeks is the standard d. ACT
d. Acceptable as long as the patient performs self-
monitoring daily using an approved home testing
instrument and reports unacceptable results promptly
to her physician
780 PART VIII Hemostasis and Thrombosis

6. What test is used most often to monitor UFH therapy in 12. What is the name of the measurable platelet activation
the central laboratory? metabolite used in the AspirinWorks assay to monitor
a. PT aspirin resistance?
b. PTT a. 11-dehydrothromboxane B2
c. ACT b. Arachidonic acid
d. Chromogenic antifactor Xa heparin assay c. Thromboxane A2
d. Cyclooxygenase
7. What test is used most often to monitor LMWH therapy
in the central laboratory? 13. Which of the following is an intravenous antiplatelet drug
a. PT used in the cardiac catheterization laboratory?
b. PTT a. Abciximab
c. ACT b. Ticlopidine
d. Chromogenic antifactor Xa heparin assay c. Prasugrel
d. Clopidogrel
8. What is an advantage of LMWH therapy over UFH therapy?
a. It is cheaper. 14. Which of the following is a newly developing oral
b. It causes less bleeding. anticoagulant?
c. It has a stable dose response. a. Argatroban
d. There is no risk of HIT. b. Lepirudin
c. Bivalirudin
9. In what situation is a DTI used? d. Rivaroxaban
a. DVT
b. HIT 15. Which of the following is not a point-of-care instrument
c. Any situation in which warfarin could be used for the measurement of PT?
d. Uncomplicated AMI a. CoaguChek XS PT
b. Gem PCL Plus
10. What laboratory test may be used to monitor DTI therapy c. Cascade POC
when PTT results are unreliable? d. Multiplate
a. PT
b. ECT
c. Reptilase clotting time
d. Chromogenic antifactor Xa heparin assay

11. What is the reference method for detecting aspirin or


clopidogrel resistance?
a. Platelet aggregometry
b. AspirinWorks
c. VerifyNow
d. PFA-100

REFERENCES
1. Schulman S, Beyth RJ, Kearon C, et al: Hemorrhagic complica- 6. Duxbury BM, Poller L: The oral anticoagulant saga: past,
tions of anticoagulant and thrombolytic treatment: American present, and future. Clin Appl Thromb Hemost 7:269-275,
College of Chest Physicians Evidence-Based Clinical Practice 2001.
Guidelines (8th edition). Chest 133:257S-298S, 2008. 7. Warkentin TE: Coumarin-induced skin necrosis and venous
2. Francis CW: Antithrombotic agents. In Kitchens CS, Alving limb gangrene. In Colman RW, Marder VJ, Clowes AW, et al,
BM, Kessler CM, editors: Consultative hemostasis and thrombo- editors: Hemostasis and thrombosis: basic principles and clinical
sis, ed 2, Philadelphia, 2007, Saunders, pp 449-460. practice, ed 5, Philadelphia, 2006, Lippincott Williams &
3. Marder VJ: Thrombolytic therapy. In Kitchens CS, Alving BM, Wilkins, pp 1663-1671.
Kessler CM, editors: Consultative hemostasis and thrombosis, 8. Ng VL: Anticoagulation monitoring. Clin Lab Med 29:
ed 2, Philadelphia, 2007, Saunders, pp 491-502. 283-304, 2009.
4. Adverse events and deaths associated with laboratory errors 9. Kitchen S, Preston FE: Standardization of prothrombin
at a hospitalPennsylvania, 2001. MMWR Morb Mortal Wkly time for laboratory control of oral anticoagulant therapy.
Rep 50:710-711, 2001. Semin Thromb Hemost 25:17-25, 1999.
5. Ansell J, Hirsh J, Hylek E, et al: Pharmacology and manage- 10. Schalekamp T, Brasse BP, Roijers JF, et al: VKORC1 and
ment of the vitamin K antagonists: American College of Chest CYP2C9 genotypes and acenocoumarol anticoagulation
Physicians Evidence-Based Clinical Practice Guidelines (8th status: interaction between both genotypes affects over-
edition). Chest 133:160S-198S, 2008. anticoagulation. Clin Pharmacol Ther 80:13-22, 2006.
CHAPTER 46 Monitoring Antithrombotic Therapies 781
11. Osinbowale O, Al Malki M, Schade A, et al: An algorithm for 31. Shantsila E, Lip GY, Chong BH: Heparin-induced thrombo-
managing warfarin resistance. Cleve Clin J Med 76:724-730, cytopenia. A contemporary clinical approach to diagnosis
2009. and management. Chest 135:1651-1664, 2009.
12. Warkentin TE, Greinacher A, Koster A, et al: Treatment 32. Cardenas GA, Deitcher SR: Risk of anaphylaxis after
and prevention of heparin-induced thrombocytopenia: re-exposure to intravenous lepirudin in patients with current
American College of Chest Physicians Evidence-Based Clini- or past heparin-induced thrombocytopenia. Mayo Clin Proc
cal Practice Guidelines (8th edition). Chest 133:340S-380S, 80:491-493, 2005.
2008. 33. Schulman S, Spencer FA: Antithrombotic drugs in coronary
13. Coagulation analyzerspoint of care, self-monitoring. CAP artery disease: risk benefit ratio and bleeding. J Thromb
Today, pp 56-60, May 2010. Haemost 8:641-650, 2010.
14. Laposata M: Point-of-care coagulation testing: stepping gently 34. Curran MP: Bivalirudin: in patients with ST-segment eleva-
forward. Clin Chem 47:801-802, 2001. tion myocardial infarction. Drugs 70:909-918, 2010.
15. Hirsch J, Bauer KA, Donati MB, et al: Parenteral anticoagu- 35. Wittkowsky AK: New oral anticoagulants: a practical guide for
lants: American College of Chest Physicians Evidence-Based clinicians. J Thromb Thrombolysis 29:182-191, 2010.
Clinical Practice Guidelines (8th edition). Chest 133:141S- 36. Chia S, Van Cott EM, Raffel OC, et al: Comparison of activated
159S, 2008. clotting times obtained using Hemochron and Medtronic
16. Olson JD, Arkin CF, Brandt JT, et al, editors: College of analyzers in patients receiving anti-thrombin therapy during
American Pathologists Conference XXXI on laboratory moni- cardiac catheterization. Thromb Haemost 101:535-540, 2009.
toring of anticoagulant therapy: laboratory monitoring of 37. Choi TS, Khan AI, Greilich PE, et al: Modified plasma-based
unfractionated heparin therapy. Arch Pathol Lab Med 122: ecarin clotting time assay for monitoring of recombinant
782-798, 1998. hirudin during cardiac surgery. Am J Clin Pathol 125:290-295,
17. Brill-Edwards P, Ginsberg JS, Johnston M, et al: Establishing 2006.
a therapeutic range for heparin therapy. Ann Intern Med 38. Lincoff AM, Tcheng JE, Califf RM, et al: For the EPILOG
119:104-109, 1993. Investigators. Sustained suppression of ischemic complica-
18. Clinical and Laboratory Standards Institute: One-stage pro- tions of coronary intervention by platelet GP IIb/IIIa block-
thrombin time (PT) test and activated partial thromboplastin time ade with abciximab. Circulation 99:1951-1958, 1999.
(APTT) test; approved guideline, ed 2, CLSI Document H27-A2, 39. CAPTURE Investigators: Randomised placebo-controlled trial
Wayne, Penn, 2008, Clinical and Laboratory Standards of abciximab before, during and after coronary intervention
Institute. in refractory unstable angina: the CAPTURE study. Lancet
19. Eikelboom JW, Hirsh J: Monitoring unfractionated heparin 349:1429-1435, 1997.
with the aPTT: time for a fresh look. Thromb Haemost 96: 40. Muhlestein JB: Effect of antiplatelet therapy on inflammatory
547-552, 2006. markers in atherothrombotic patients. Thromb Haemost
20. Tripodi A, van den Besselaar A: Laboratory monitoring of 103:71-82, 2010.
anticoagulation: where do we stand? Semin Thromb Hemost 41. vant Hof AW, Valgimigli M: Defining the role of platelet
35:34-41, 2009. glycoprotein receptor inhibitors in STEMI: focus on tirofiban.
21. Bharadwaj J, Jayaraman C, Shrivastava R: Heparin resistance. Drugs 69:85-100, 2009.
Lab Hematol 9:125-131, 2003. 42. Gilroy DW: Eicosanoids and the endogenous control of acute
22. Adcock DM, Kressin DC, Marlar RA: The effect of time and inflammatory resolution. Int J Biochem Cell Biol 42:524-528,
temperature variables on routine coagulation tests. Blood 2010.
Coagul Fibrinolysis 9:463-470, 1998. 43. Juul-Moller S, Edvardsson N, Jahnmatz B, et al: Double-blind
23. Estry DW, Wright L: Laboratory assessment of anticoagulant trial of aspirin in primary prevention of myocardial infarction
therapy. Clin Lab Sci 1:161-164, 1988. in patients with stable chronic angina pectoris. Lancet
24. Slight RD, Buell R, Nzewi OC, et al: A comparison of acti- 340:1421-1425, 1992.
vated coagulation timebased techniques for anticoagulation 44. The RISC Group: Risk of myocardial infarction and death
during cardiac surgery with cardiopulmonary bypass. J Car- during treatment with low dose aspirin and intravenous
diothorac Vasc Anesth 22:47-52, 2008. heparin in men with unstable coronary artery disease. Lancet
25. The perioperative management of antithrombotic therapy: 336:827-830, 1990.
American College of Chest Physicians Evidence-Based Clini- 45. ISIS-2 Collaborative Group: Randomised trial of intravenous
cal Practice Guidelines (8th edition). Chest 133:299S-339S, streptokinase, oral aspirin, both or neither among 17,187
2008. cases of suspected acute myocardial infarction: ISIS-2. Lancet
26. McGlasson DL, Kaczor DA, Krasuski RA, et al: Effects of 2:349-360, 1988.
pre-analytical variables on the antiactivated factor X chro- 46. The Dutch TIA Study Group: A comparison of two doses of
mogenic assay when monitoring unfractionated heparin and aspirin (30mg vs. 283mg a day) in patients after a transient
low molecular weight heparin anticoagulation. Blood Coagul ischemic attack or minor ischemic stroke. N Engl J Med
Fibrinolysis 16:173-176, 2005. 325:1261-1622, 1991.
27. McGlasson DL: Using a single calibration curve with the 47. Hirsh J, Dalen JE, Fuster V, et al: Aspirin and other platelet-
anti-Xa chromogenic assay for monitoring heparin antico- active drugs: the relationship among dose, effectiveness, and
agulation. Lab Med 36:297-299, 2005. side effects. Chest 108:247S-257S, 1995.
28. Samama MM, Gerotziafas GT: Newer anticoagulants in 2009. 48. International Stroke Trial Collaborative Group: The Interna-
J Thromb Thrombolysis 29:92-104, 2010. tional Stroke Trial (IST): a randomised trial of aspirin, sub-
29. Fuji T, Fujita S, Tachibana S, et al: Randomized, double-blind, cutaneous heparin, both, or neither among 19,435 patients
multi-dose efficacy, safety and biomarker study of the oral with acute ischemic stroke. Lancet 349:1569-1581, 1997.
factor Xa inhibitor DU-176b compared with placebo for 49. CAST (Chinese Acute Stroke Trial) Collaborative Group:
prevention of venous thromboembolism in patients after CAST: randomised placebo-controlled trial of early aspirin
total knee arthroplasty. ASH Annual Meeting Abstracts. use in 20,000 patients with acute ischemic stroke. Lancet
Blood 112:34, 2008. 349:1641-1649, 1997.
30. Yeh RW, Jang IK: Argatroban: update. Am Heart J 151: 50. Antiplatelet Trialists Collaboration: Collaborative overview
1131-1138, 2006. of randomized trials of antiplatelet therapy: I. prevention of
782 PART VIII Hemostasis and Thrombosis

death, myocardial infarction, and stroke by prolonged 58. Gum PA, Kottke-Marchant K, Welsh PA, et al: A prospective,
antiplatelet therapy in various categories of patients. BMJ blinded determination of the natural history of aspirin resis-
308:81-106, 1994. tance among stable patients with cardiovascular disease. J Am
51. Hennekens CH, Peto R, Hutchison GB, et al: An overview Coll Cardiol 41:961-965, 2003.
of the British and American aspirin studies. N Engl J Med 59. Chen WH: Antiplatelet resistance with aspirin and clopido-
318:923-924, 1988. grel: is it real and does it matter? Curr Cardiol Rep 8:301-306,
52. Manson JE, Stampfer MJ, Colditz GA, et al: A prospective 2006.
study of aspirin use and primary prevention of cardiovascular 60. Knoepp SM, Laposata M: Aspirin resistance: moving forward
disease in women. JAMA 266:521-527, 1991. with multiple definitions, different assays, and a clinical
53. Fitzgerald GA, Meagher EA: Antiplatelet drugs. Eur J Clin imperative. Am J Clin Pathol 123:S125-S132, 2005.
Invest 24(suppl 1):46-49, 1994. 61. van Werkum JW, Harmsze AM, Elsenberg EH, et al: The
54. Freeman MK: Thienopyridine antiplatelet agents: focus on use of the VerifyNow system to monitor antiplatelet therapy:
prasugrel. Consult Pharm 25:241-257, 2010. a review of the current evidence. Platelets 19:479-488,
55. Mousa SA, Jeske WP, Fareed J: Antiplatelet therapy prasugrel: 2008.
a novel platelet ADP P2Y12 receptor antagonist. Clin Appl 62. Mueller T, Dieplinger B, Poelz W, et al: Utility of the PFA-100
Thromb Hemost 16:170-176, 2010. instrument and the novel Multiplate analyzer for the assess-
56. Helgason CM, Bolin KM, Hoff JA, et al: Development of ment of aspirin and clopidogrel effects on platelet function
aspirin resistance in persons with previous ischemic stroke. in patients with cardiovascular disease. Clin Appl Thromb
Stroke 25:2331-2336, 1994. Hemost 15:652-659, 2009.
57. Eikelboom JW, Hirsh J, Weitz JI, et al: Aspirin-resistant throm- 63. McGlasson DL, Chen M, Fritsma GA: Urinary 11-
boxane biosynthesis and the risk of myocardial infarction, dehydrothromboxane B2 levels in healthy individuals follow-
stroke, or cardiovascular death in patients at high risk for ing a single dose response to two concentrations of aspirin.
cardiovascular events. Circulation 105:1650-1655, 2002. J Clin Ligand Assay 28:147-150, 2005.
Coagulation Instrumentation*
David L. McGlasson
47
OUTLINE OBJECTIVES
Historical Perspective After completion of this chapter, the reader will be able to:
Clot End-Point Detection
Principles 1. Describe testing methodologies previously con 7. Identify key performance characteristics that should
Electromechanical sidered as specialized that are now routinely avail be evaluated when selecting the most appropriate
End-Point Detection able on coagulation analyzers. coagulation analyzer for an individual laboratory
Photo-optical End-Point 2. Identify testing applications for various coagulation setting.
Detection analyzers. 8. Explain the purpose of incorporating platelet function
Nephelometric End-Point 3. Explain the methods of clot detection used by each testing analyzers into the routine coagulation
Detection type of coagulation analyzer presented. laboratory.
Chromogenic End-Point 4. List common instrument flags that alert operators to 9. Develop a model plan of action for objective evalua
Detection specimen and instrument problems. tion of coagulation analyzers for purchase.
Immunologic Light
5. Describe the advantages and disadvantages of each 10. Explain the main purpose of point-of-care coagula
Absorbance End-Point
method of clot detection. tion testing.
Detection
Advances in 6. Distinguish the characteristics of manual, semiauto
Coagulometry mated, and automated coagulation analyzers.
Improved Accuracy and
Precision
CASE STUDY
Random Access Testing
Improved Reagent After studying the material in this chapter, the reader should be able to respond to the following
Handling case study:
Improved Specimen
Management A 35-year-old white man was admitted to the hospital with abdominal pain and tenderness,
Expanded Computer malaise, and a low-grade fever. A tentative diagnosis of cholecystitis was made, with possible surgi-
Capabilities cal intervention considered. Pertinent medical history included tonsillectomy at age 6 and appen-
Other Automated dectomy at age 18 with no abnormal bleeding symptoms noted. The patient reported that he was
Features taking no medications at this time. In anticipation of surgery, routine coagulation studies were
Specimen Quality Flags ordered. When the specimen arrived in the laboratory, it was centrifuged, and it was noted that the
Instrument Malfunction plasma had a whitish, milky appearance. The specimen was processed by an automated analyzer
Flags using photo-optical end-point detection methodology, and the following results were obtained:
Advantages and
Disadvantages of Test Results Reference Interval Flags
End-Point Detection Prothrombin time 16.7sec 10.9-13.0sec Lipemia
Methods
Partial thromboplastin time >150sec 30.6-35.0sec Lipemia
Platelet Function Testing
Fibrinogen 245mg/dL 190-410mg/dL Lipemia
Instruments
Point-of-Care Testing Because the laboratorys policy is to retest after all abnormal coagulation results, the prothrom-
Selection of Coagulation bin time and partial thromboplastin time assays were repeated, and similar values were obtained.
Instrumentation 1. Should the operator report the test results as shown?
Currently Available
2. What action should the operator take to address the lipemia flagging of this specimen?
Instruments
3. Would you expect this patient to be at risk for bleeding based on these test results?

*All chapter contents constitute the personal statements of the author, and no endorsement by the U.S. Air Force or any other federal government
entity is intended or implied.

783
784 PART VIII Hemostasis and Thrombosis

T
he coagulation laboratory is an ever-changing environ- the groundwork for the most commonly ordered coagulation
ment populated by automated analyzers that offer tests in current use, the PT and PTT.3 Subsequent twentieth-
advances in both volume and variety of tests.1 Hardware century developments include manual loops, electromechani-
and software innovations provide for random access testing cal clot detection using a movable lead (BBL Fibrometer) or
with multitest profiles. In the past, the routine coagulation rolling steel ball (Diagnostica Stago ST-4), and photo-optical
test menu consisted of prothrombin time (PT) with the clot detection (Coag-A-Mate, originally manufactured by
international normalized ratio (INR), partial thromboplastin General Diagnostics, Chapel Hill, N.C.). Nephelometers, first
time (PTT, activated partial thromboplastin time), fibrinogen, developed in 1920, measure 90-degree light dispersion in a
thrombin time, and D-dimer assays. More specialized testing colloidal suspension. As plasma clots, a change in light scatter
was performed in tertiary care institutions or reference labora- can be measured over time, and this principle is in common
tories employing medical laboratory scientists with specialized use today.
training. With the introduction of new instrumentation The 1950s saw the development of the BBL Fibrometer, an
and test methodologies, coagulation testing capabilities have instrument that can still be found in coagulation laboratories,
expanded significantly, so that many formerly specialized although it is no longer being manufactured. This instrument
tests can be performed easily by general medical laboratory employed an electromechanical clot detection methodology
staff. New instrumentation has made coagulation testing more that allowed laboratories to transition from the manual tilt
standardized, consistent, and cost effective. Automation has tube or wire loop method to a more accurate semiautomated
not advanced, however, to the point of making coagulation testing process.
testing foolproof or an exact science. Operators must develop Current coagulation instruments apply many of the clot
expertise in correlating critical test results with the patients detection principles of these early analytical systems. They either
diagnosis or condition when monitoring antithrombotic observe for the clot formation (optical, nephelometric devices)
therapy. Good method validation of procedures, cognitive or they detect the clot by feel (mechanical, viscosity-based
ability, and theoretical understanding of the hemostatic mech- devices). Although the principles remain the same, the instru-
anism are still required to ensure the accuracy and validity of ments have been enhanced to eliminate variations in pipetting
test results so that the physician can make an informed deci- and end-point detection. They also allow multiple analyses to
sion about patient care. be performed simultaneously on a single specimen.4

HISTORICAL PERSPECTIVE CLOT END-POINT DETECTION PRINCIPLES


Visual clot-based testing began in the eighteenth century. Labo- Coagulometers are automated or semiautomated. Semiauto-
ratorians worked with animals and human blood, observing mated coagulometers require the operator to deliver test plasma
how long it took blood to clot after collection. With the advent and reagents manually to the reaction cuvette and limit testing
of the microscope in the 1800s scientists observed blood to to one or two specimens at a time. They are relatively inexpen-
look for increasing turbidity and visible clot formation. sive, but their use requires considerable operator expertise.
Many advancements took place from 1822 to 1921. These Fully automated analyzers provide pipetting systems that
included controlling the temperature during the formation of automatically transfer reagents and test plasma to reaction
a clot, passing objects through the blood to detect resistance, vessels and measure the end point without operator interven-
and using glass capillary tubes to view clot formation. In the tion (Table 47-1). Multiple specimens can be tested simultane-
early 1900s, researchers monitored the length of time it ously. Automated coagulometers are expensive, and laboratory
took whole blood to clot in glass tubes while they were being staff require specialized training to operate and maintain them.
tilted, a precursor to the Lee-White tilt tube procedure of sub- Regardless of technology, all semiautomated and automated
sequent years. analyzers offer better coagulation testing accuracy and preci-
The coaguloviscosimeter, a primitive whole-blood clot sion than visual methods.
detection device, was developed in 1910. The change in vis Instrument methodologies are classified into five groups
cosity as blood clotted generated a voltage change that was based on the end-point detection principle:
recorded by a direct readout system. Voltage changes could be Mechanical
plotted against time to measure clot formation. Photo-optical (turbidometric)
Except for point-of-care testing and platelet aggregometry, Nephelometric
citrated plasma (usually platelet-poor plasma, plasma with a Chromogenic (amidolytic)
platelet count of less than 10,000/mcL) has now replaced Immunologic
whole blood in coagulation instruments, although the princi- Historically, coagulation instruments were limited to a
ple of interval to clot formation lives on.1,2 single type of end-point detection system each, mechanical or
Plasma coagulation testing traces its roots to Gram, who photo-optical. Photo-optical instruments operated at a fixed
in 1920 added calcium chloride to anticoagulated plasma wavelength between 500nm and 600nm and could be used
at 37 C. He measured the increasing viscosity of the blood only for clot detection. Later instruments could also read at
during fibrin monomer polymerization, a principle used today 405nm to perform chromogenic assays. Although this change
in thromboelastography (TEG) and sonar clot detection, laying made it possible to automate advanced coagulation protocols,
CHAPTER 47 Coagulation Instrumentation 785

TABLE 47-1 Levels of Coagulation Automation


Level Description Examples
Manual All reagents and specimens are transferred manually by the operator. Tilt tube
Temperature is maintained by a water bath or heat block; external Wire loop
measurement by operator may be required. End point is determined
visually by the operator. Timer is initiated and stopped by the operator.
Semiautomated All reagents and specimens are transferred manually by the operator. Fibrometer
Instrument usually contains a device for maintaining constant 37 C STart 4
temperature. Analyzer may internally monitor temperature. Instrument Cascade M-4
has mechanism to initiate timing device automatically on addition of Cascade-M
final reagent and mechanism for detecting clot formation and stopping BFT-II
the timer. KC1
KC4
Automated All reagents are automatically pipetted by the instrument. Specimens may CoaLab
or may not be automatically pipetted. Analyzer contains monitoring STA-R Evolution
devices and internal mechanism to maintain and monitor constant STA Compact and Compact CT
37 C temperature throughout testing sequence. Timers are initiated Sysmex CA-CA-530, CA-560, CA-1500, CA-7000
and clot formation is detected automatically. BCS XP

it required laboratories to purchase and train staff on multiple


analyzers if they wanted to offer the wider range of coagulation
testing capabilities. Since 1990, instrument manufacturers have
successfully incorporated multiple detection methods into
single analyzers, which allows a laboratory to purchase and
train on only one instrument while still providing specialized
testing capabilities implemented by multiple operators.4

Electromechanical End-Point Detection


Amplitude

In electromechanical clot detection systems such as the system


employed by BBLs time-honored Fibrometer, fibrin strands
attach to a moving mechanical electrode (probe), completing
an electrical circuit and stopping the interval timer. There is Amount of clotting
one stationary and one moving probe. During clotting, the Figure 47-1 Viscosimetric (electromechanical) clot detection in a Diag
moving probe enters and leaves the plasma at regular intervals. nostica Stago analyzer. A steel ball oscillates in an arc from one side of the
The current between the probes is broken as the moving probe cuvette to the other. Movement is monitored continuously within a magnetic
leaves the plasma. When a clot forms, the fibrin strand con- field. As the sample clots, viscosity rises and movement of the steel ball is
ducts current between the probes even when the moving probe impeded. Variation in amplitude stops the timer, and the interval is the
exits the solution. The current completes a circuit and stops clotting time.
the timer.
Another mechanical clot detection method employs a mag-
netic sensor that monitors the movement of a steel ball within and Destiny instruments distributed by Tcoag US, a division
the test plasma. Two moving ball principles are used. In the of Diagnostica Stago located in Parsippany, N.J. Mechanical
first, an electromagnetic field detects the oscillation of a steel methods are unaffected by plasma icterus or lipemia.
ball within the plasma-reagent solution. As fibrin strands form,
the viscosity starts to increase, slowing the movement (Figure Photo-optical End-Point Detection
47-1). When the oscillation decreases to a predefined rate, the Photo-optical (turbidometric) coagulometers detect a change
timer stops, indicating the clotting time of the plasma. This in plasma optical density (OD, transmittance) during clotting.
methodology is found on all Diagnostica Stago analyzers. Light of a specified wavelength passes through plasma, and its
In the second variation, a steel ball is positioned in an intensity (OD) is recorded by a photodetector. The OD depends
inclined well. The position of the ball is detected by a magnetic on the color and clarity of the sample and is established as the
sensor. As the well rotates, the ball remains positioned on the baseline. Formation of fibrin strands causes light to scatter,
incline. When fibrin forms, the ball is swept out of position. allowing less to fall on the photodetector and thus generating
As it moves away from the sensor, there is a break in the circuit, an increase in OD. When the OD rises to a predetermined vari-
which stops the timer. This technology can be found on AMAX ance from baseline, the timer stops, indicating clot formation.
786 PART VIII Hemostasis and Thrombosis

Collim. lens Interferential Nephelometry can be adapted to dynamic clot measure-


Lamp filter ment. Continuous readings are taken throughout clotting,
measuring the entire clotting sequence to completion and pro-
ducing a clot curve or signature.
Nephelometry was first applied to immunoassay. As antigen-
Fiber optics antibody complexes (immune complexes) precipitate, the
resulting turbidity scatters incident light.6 In reactions in which
the immune complexes are known to be too small for detec-
tion, the antibodies are first attached to microlatex particles.
In coagulation, coagulation factor immunoassays employ
specific factor antibodies bound to latex particles. Nephelom-
Lens etry provides a quantitative, but not functional, assay of coagu-
Diaphram Curvette Sensor lation factors. Nephelometry is often employed in complex
Figure 47-2 Photo-optical (turbidometric) clot detection. Polychromatic automated coagulometers because it allows both clot-based
light is focused by a collimator and filtered to transmit a selected wavelength. assay and immunoassay. Nephelometry-style analyzers can
Monochromatic light is transmitted by fiber optics and focused on the reac be used to produce high-volume multiple-assay coagulation
tion cuvette. As fibrin forms, opacity increases and the intensity of light profiles.
reaching the sensor decreases. Collim., Collimator.
Chromogenic End-Point Detection
Chromogenic (synthetic, amidolytic) methodology employs a
colorless synthetic oligopeptide substrate conjugated to a chro-
mophore, usually para-nitroaniline (pNA). Chromogenic anal-
ysis is a means for measuring specific coagulation factor activity,
because it exploits the factors enzymatic (protease) properties.
Sensor The oligopeptide is a series of amino acids whose sequence
matches the natural substrate of the protease being measured.7
Protease cleaves the chromogenic substrate at the site binding
the oligopeptide to the pNA, freeing the pNA. Free pNA is
yellow; the OD of the solution is proportional to protease
activity and is measured by a photodetector at 405nm. In
some instances a fluorescent conjugate is used; this method is
Light
source called fluorogenic.
A B
The activity of coagulation factors and many other
Figure 47-3 Nephelometric clot detection. A, Light from below passes
enzymes is measured by the chromogenic method directly or
through the sample to the detector above. B, As fibrin polymerizes, light is
deflected and is detected at an angle from the original path. indirectly:
Direct chromogenic measurement: OD is proportional to the
activity of the substance being measured, for instance,
coagulation control protein C activity.
Indirect chromogenic measurement: The protein or analyte
Because the baseline OD is subtracted from final OD, effects of being measured inhibits a reagent enzyme that has activity
lipemia and icterus are minimized. Many optical systems directed toward the synthetic substrate. The change in OD
employ multiple wavelengths that discriminate and filter out is inversely proportional to the concentration or activity
the effects of icterus and lipemia. Many automated and semi- of the substance being measured; for example, heparin in
automated coagulation instruments developed since 1970 use the antifactor Xa assay. The principle used in instruments
photo-optical clot detection (Figure 47-2). marketed by the DiaPharma Group, Inc. (West Chester,
Ohio) to perform chromogenic factor X assay is shown in
Figure 47-4.
Nephelometric End-Point Detection
Nephelometry is a modification of photo-optical end-point Immunologic Light Absorbance
detection in which 90-degree or forward-angle scatter, rather End-Point Detection
than OD, is measured. A light-emitting diode produces inci- Immunologic assays are the newest assays available in coagula-
dent light at approximately 600nm and a photodetector tion laboratories and are based on antigen-antibody reactions
detects variations in light scatter at 90 degrees (side scatter) and similar to those used in nephelometry as described previously.
at near 180 degrees (forward-angle scatter). As fibrin polymers Latex microparticles are coated with antibodies directed against
form, side scatter and forward-angle scatter rise (Figure 47-3).4,5 the selected analyte (antigen). Monochromatic light passes
The timer stops when scatter reaches a predetermined intensity, through the suspension of latex microparticles. When the
and the interval is recorded. wavelength is greater than the diameter of the particles, only a
CHAPTER 47 Coagulation Instrumentation 787

RVV processing, increased throughput, and enhanced data manage-


1 FX FXa
Ca2 ment and result traceability.

Improved Accuracy and Precision


FXa In the days of visual methods, coagulation assays were per-
2 FXa-specific substrate Peptide  pNA formed in duplicate to reduce the coefficient of variation,
Figure 47-4 Method used in the DiaPharma chromogenic factor X assay. which generally exceeded 20%. Semiautomated instruments
The assay may be a useful tool in the management of patients with lupus have improved upon precision, but the requirement for manual
inhibitors who are receiving warfarin or direct thrombin inhibitors. It also pipetting of plasma and reagents continues to necessitate
may be used when the conventional prothrombin time/international normal duplicate testing. With the advent of fully automated instru-
ized ratio testing is not suitable for individual patients. The method involves
ments, precision has improved to the extent that single testing
two stages. In stage 1, the activator Russell viper venom (RVV) activates
can be performed with confidence, halving material and
factor X (FX) in the presence of calcium to factor Xa (FXa). In stage 2, the
generated FXa hydrolyzes the chromogenic substrate S-2765, liberating the reagent costs. Coefficients of variation of less than 5%, and
chromophore group para-nitroaniline (pNA). The color is then read photo even less than 1% for some tests, have been achieved. Initial
metrically at 405nm. The amount of FXa generated, and thus the intensity accuracy and precision are established by method validation
of the color, is proportional to the FX activity in the sample over the assay for all instrument and reagent combinations (see Chapter 5).
range.
Random Access Testing
Automated coagulometers now provide random access testing.
small amount of light is absorbed.8 When the coated latex Through simple programming, a variety of tests can be run in
microparticles come into contact with their antigen, however, any order on single or multiple specimens within a testing
the antigen attaches to the antibody and forms bridges, sequence. Previous automated analyzers were capable of
which causes the particles to agglutinate. As the diameter of the running only one or two assays at a time, so batching was
agglutinates increases to the wavelength of the monochromatic necessary. The disadvantage was that specimens with multiple
light beam, light is absorbed. The increase in light absorbance orders had to be handled multiple times. For current automated
is proportional to the size of the agglutinates, which in turn is analyzers, the ability to run multiple tests is limited only by
proportional to the antigen level. the number of reagents that can be stored in the analyzer and
Immunoassay technology became available on coagulom- the instruments ability to interleave tests requiring different
eters in the 1990s and is used to measure a growing number end-point detection methodologies simultaneously, such as
of coagulation factors and proteins, expanding the diagnostic clot-based, chromogenic, and immunologic methods. Random
capabilities of the laboratory. Coagulation protein testing, access promotes profiling.
which used to take hours or days to perform using traditional
antigen-antibody detection methodologies such as enzyme- Improved Reagent Handling
linked immunosorbent assay or electrophoresis, now can be Reduced Reagent and Specimen Volumes
done in minutes on an automated analyzer. Automated and semiautomated coagulometers now have the
The introduction of new coagulation methodologies has capability to perform tests on smaller sample volumes. Tradi-
improved testing capabilities in the coagulation laboratory. tionally, PT assays required 0.1mL of patient plasma and
Refinement of these methodologies has allowed the use of 0.2mL of thromboplastin/calcium chloride reagent. PTT was
synthetic substrates and measurements of single proenzymes, measured using 0.1mL of plasma, 0.1mL of activated partial
enzymes, and monoclonal antibodies, which increases the thromboplastin, and 0.1mL of calcium chloride. Current ana-
ability to recognize the causes of disorders of hemostasis and lyzers can perform the same tests using one half or even one
thrombosis.8 quarter the traditional volumes of reagents and patient speci-
mens. This promotes the use of smaller specimen volumes,
especially from pediatric patients or those from whom speci-
ADVANCES IN COAGULOMETRY
mens are difficult to draw, and further reduces reagent costs.
Significant advances have been made in the capability and
flexibility of coagulation instrumentation. Instruments previ- Open Reagent Systems
ously required manual pipetting, recording, and calculation of A variety of reagents from numerous distributors are available
results, which necessitated significant operator expertise, inter- for coagulation testing, and laboratory directors want the flex-
vention, and time. Current technology allows a walkaway ibility of selecting the reagents that best suit their needs without
environment in which, after specimens and reagents are loaded being restricted in their choices by the analyzers being used.
and the testing sequence is initiated, the operator can move on Recognizing that the ability to select reagents independently of
to perform other tasks. the test system is a high priority, instrument manufacturers
Clot detection methods have remained consistent, and have responded by developing systems that provide optimal
multiple methodologies have been incorporated into single performance with alternative manufacturers reagents, pro-
analyzers to expand their test menu options. Advances vided that the reagents are compatible with the instruments
have included improved specimen and reagent storage and methodology.
788 PART VIII Hemostasis and Thrombosis

Reagent Tracking control files can be stored, which eliminates the time-
Many automated instruments keep records of reagent lot consuming task of manually logging and graphing quality
numbers and expiration dates, which makes it easier for the control values. Westgard rules (see Chapter 5) can be applied,
laboratory to maintain reagent integrity and comply with regu- and failures are automatically flagged. Some analyzers feature
latory requirements. Additional features often include on-board automatic repeat testing when failures occur on the initial run.
monitoring of reagent volumes with flagging systems to alert The quality control files can be reviewed or printed on a regular
the operator when an insufficient volume of reagent is present basis to meet regulatory requirements.
in relation to the number of specimens programmed to be run. The programming flexibility of modern analyzers has
Reagent bar coding supports record keeping because it tracks enhanced the laboratorys opportunities to provide expanded
reagent properties and enables the operator to load coagulo test menus. Most advanced analyzers are preprogrammed with
meters without stopping specimen analyses. several routine test protocols ready for use. Specimen and
reagent volumes, incubation times, and other testing parame-
Improved Specimen Management ters do not need to be predetermined by the operator but can
Primary Tube Sampling be changed easily when necessary. Additional tests can be pro-
Many coagulometers encourage the operator to place the grammed into the analyzer by the user whenever needed, which
primary collection tube on the instrument after centrifugation, allows for enhanced flexibility of the analyzer and reduces the
which eliminates the need to separate the plasma into a need for laboratories to have multiple instruments.
secondary tube. In addition, instruments often accommodate Instrument interfacing to laboratory information systems
multiple tube sizes. Significant time savings occur as a result of and specimen bar coding capabilities have become a priority
elimination of the extra specimen preparation step, and errors as facilities of all sizes endeavor to reduce dependence on
resulting from mislabeling of the aliquot tube are reduced. manual record keeping. Bidirectional interfaces improve effi-
ciency through the ability of the instrument to send specimen
Closed-Tube Sampling bar code information to the laboratory information systems
Closed-tube sampling has improved the safety and efficiency and receive a response listing the tests that have been ordered.
of coagulation testing. After centrifugation, the tube is placed This eliminates the need for the operator to program each
on the analyzer without removing the blue stopper. The cap is specimen and test.
pierced by a needle that aspirates plasma without disturbing
the red blood cell layer. Not only does closed-tube sampling Other Automated Features
save staff time, it also reduces the risk of specimen exposure A few additional features offered by current coagulation ana-
through aerosols or spillage. Closure also promotes plasma pH lyzers should be mentioned.
stabilization. When closed-tube sampling is used, specimens Improved flagging capabilities alert the operator when preset
are visually checked for clots after centrifugation or even after criteria have been exceeded (Box 47-1). Flags may indicate
analysis. instrument malfunction such as cuvette jams, low reagent
volume, and temperature errors, or a problem with the
Flagging for Specimen Interferences results such as values that exceed critical limits, inability to
Some analyzers monitor the quality of the test specimen for detect an accurate end point, or values outside of the linear
interfering substances or unusual testing characteristics, such range.
as hemolysis, lipemia, bilirubinemia (icterus), abnormal clot- Reflex testing is the automatic ordering of tests based on
ting patterns, or results that fall outside the linear range of the preset parameters or the results of prior tests. Instruments
reference curve (values above the top curve point or below the may make additional dilutions if the initial result is outside
bottom curve point). Flags warn the operator of potential
errors so that problems can be resolved in a timely manner
(see later).
BOX 47-1 Warning Flags Available on Coagulometers
Automatic Dilutions
Instrument Malfunction Flags
Many instruments perform multiple dilutions on patient speci-
Temperature error
mens, calibrators, or controls, eliminating the need for the
Photo-optics error
operator to perform this task manually and reducing the poten-
Mechanical movement error
tial for dilution errors. These conditions can be automatically
Probe not aspirating
programmed into the individual test setups on the analyzers
being used.
Sample Quality Flags
Lipemia
Expanded Computer Capabilities Hemolysis
The computer circuitry of analyzers now incorporates internal
Icterus
data storage and retrieval systems. Hundreds of results can be
Abnormal clot formation
stored, retrieved, and compiled into cumulative reports. Mul-
No end point detected
tiple calibration curves can be stored and accessed. Quality
CHAPTER 47 Coagulation Instrumentation 789

No end point detected: indicates that the instrument was


144 Deceleration Endpoint unable to detect clot formation; the specimen may need to
128 be tested using an alternate methodology.
112
Instrument Malfunction Flags
mAbs

96
Instrument malfunction flags are as follows:
80
Temperature error
64 Photo-optics error
48 Mechanical movement error
Acceleration
Baseline Probe not aspirating
32
No end point detected
0 14 28 42 56 70 84 98 112
Specimen track error
Figure 47-5 Clot signature produced by the ACL TOP analyzer. The clot
curve consists of baseline, acceleration phase, deceleration phase, and end
point. The baseline is recorded before clotting occurs. Acceleration reflects ADVANTAGES AND DISADVANTAGES OF
clotting and is normally steep, because clotting is rapid. The deceleration END-POINT DETECTION METHODS
phase represents the decreasing rate of clot formation as all available fibrino
gen converts to fibrin. The end point is flat and stable, reflecting consumption Photo-optical end-point detection may be confounded by
of all fibrinogen. A key component in evaluating the clot curve is the y-axis, icterus or lipemia, which erroneously prolongs the clotting
absorbance, which provides the clot interval when it reaches a defined time, because the change in OD is masked by the color or
minimum. Absorbance adjusts to compensate for baseline fibrinogen and turbidity of the specimen. Some coagulation instruments may
interferences such as lipemia or icterus. (Photo courtesy Beckman Coulter, be unable to employ synthetic reagents because they are more
Inc., Brea, Calif.) translucent.9 Table 47-2 summarizes the advantages and dis
advantages of each end-point detection methodology.

of the linearity limits, or supplementary tests can be run


PLATELET FUNCTION
automatically if clinically indicated by the initial test result.
TESTING INSTRUMENTS
The first result does not need to wait for review by the opera-
tor before follow-up action is taken. The demand for rapid, cost-effective methods for the evalua-
Graphing of clot formation is provided on analyzers such as tion of platelet function has been rising because of the need to
the ACL TOP (Beckman Coulter). The graph is generated by monitor the efficacy of therapy with aspirin, clopidogrel, and
an algorithm, a formula used to convert raw optical mea- platelet membrane glycoprotein IIb/IIIa inhibitors (see Chapter
surements into a clotting time. Besides determining the clot- 46) for prevention of cardiovascular disease. In addition, pre-
ting time, it also smooths the raw data into a visible curve operative evaluation of platelet function is crucial in hemo-
and uses the curve to check for clot integrity. Multiple checks static management, particularly if the patient has a history of
are performed to ensure an accurate and reproducible result. bleeding.10 Platelet function testing always has been a challenge
Should the data not meet all of the acceptable criteria, an for the clinical laboratory because of the lack of reliable, accu-
error flag is generated. The clot curve is examined to trouble- rate, and easy-to-perform methods. Platelet function histori-
shoot potential technical aberrations. cally has been assessed by the Mielke bleeding time assay,
The clot formation graph may also be used as a signature which is technically demanding and fails to correlate with
that correlates with disease state. Figure 47-5 shows an intraoperative bleeding risk.11-15 Platelet aggregometry, includ-
example of a typical clot curve. ing light-transmittance and whole-blood aggregometry and
lumiaggregometry (see Chapter 45), is confined to tertiary care
Specimen Quality Flags and specialty hemostasis laboratories because it requires
Specimen quality flags include the following: advanced operator skills. Simplified and point-of-care platelet
Lipemia: may cause falsely prolonged clotting times function instrumentation is now available.
on OD instruments because of interference with light The PFA-100 platelet function analyzer (Siemens Healthcare
transmittance. Diagnostics, Inc., Deerfield, Ill.) employs blood flow and shear
Hemolysis: may cause falsely shortened clotting times rate to assess platelet function.15 Test cartridges contain mem-
because of premature activation of coagulation factors and branes coated with collagen/epinephrine or collagen/adenosine
platelets. diphosphate to stimulate platelet aggregation. Whole blood is
Icterus (bilirubinemia): indicates liver dysfunction that may aspirated under controlled flow conditions through a micro-
lead to prolonged clotting times because of inadequate scopic aperture in the membrane. The time required for a
clotting factor production; also may interfere with OD platelet plug to occlude the aperture is an indicator of platelet
instruments. function.15 The PFA-100 system is successful at detecting von
Abnormal clot formation: may lead to falsely elevated clotting Willebrand disease and the efficacy of aspirin therapy.16,17
times because of instrument inability to detect an end The Accumetrics VerifyNow system (San Diego, Calif.) is
point. an optical detection system that measures platelet-induced
790 PART VIII Hemostasis and Thrombosis

TABLE 47-2 Advantages and Disadvantages of End-Point Detection Systems


End-Point Detection Method Advantages Disadvantages
Mechanical No interference from specimen lipemia or Inability to observe graph of clot formation
bilirubinemia (icterus)
Ability to use specimen and reagent volumes as
small as 25mcL in some instruments
Photo-optical Ability to observe graph of clot formation with Interference from lipemia, hemolysis, bilirubinemia, and
some instrumentation increased plasma proteins; this issue has been
Good precision addressed by some manufacturers with readings from
Increased test menu flexibility and specimen multiple wavelengths
quality information when multiple wavelengths May not detect short clotting times owing to long lag phase
are used May not detect small friable clots that are translucent
Chromogenic Ability to measure proteins that do not clot Limited by wavelength capabilities of instrument
Expanded menu options to replace clottable assays May need large test volume to be cost effective
affected by preanalytical variables such as
heparin use or interference by direct thrombin
inhibitors such as argatroban or hirudin
Most automated systems now have cost-effective
chromogenic capabilities
Immunologic Ability to automate tests previously available only Limited number of automated tests available
with manual, time-consuming methods, such as Higher cost of instruments and reagents
enzyme-linked immunosorbent assay May need to have additional instruments available to run
Expanded test menu capabilities routine tests in laboratories without automated
coagulation analyzers that have random access capability
Nephelometric Ability to measure antigen-antibody reactions for Limited number of tests available
proteins present in small concentrations Higher cost of reagents
Need for special staff training

aggregation by microbead agglutination. The system employs frequency of 0.1Hz. In the cup, suspended by a torsion wire,
a disposable cartridge that contains lyophilized fibrinogen- is a stationary pin whose diameter is 1mm smaller than that
coated beads and a platelet agonist specific for the test. Whole of the cup.
blood is automatically dispensed from the blood collection Kaolin is added to trigger clotting. As the blood clots,
tube into the assay device, with no blood handling required fibrin links the pin to the cup and viscoelasticity changes are
by the operator. The instrument provides a Food and Drug transmitted to the pin. The resulting pin torque generates an
Administration (FDA)approved aspirin assay using arachi- electrical signal from the torsion wire that is plotted as a
donic acid as the agonist, a glycoprotein IIb/IIIa inhibitor function of time to produce a TEG tracing (Figure 47-6).
(abciximab, tirofiban, eptifibatide) assay using thrombin The tracing is analyzed to determine the speed, strength, and
receptor activation peptide (TRAP) as the agonist, and a stability of clot formation and the downstream effect of
P2Y12 inhibitor (clopidogrel, prasugrel) assay using adenosine fibrinolysis. Viscoelasticity depends on procoagulant activity,
diphosphate (ADP).18-21 cellular components (red blood cells, white blood cells,
The TEG Thromboelastograph Hemostasis Analyzer System and platelets), and fibrinolysis. The trace furnishes real-
(Haemoscope Corporation, Niles, Ill., a division of Haemonet- time information about the evolving clot from platelet
ics Corporation) is an operator-dependent system that pro- activation to initial fibrin formation, fibrin cross-linkage, and
vides global hemostasis assessment. The operator uses TEG to fibrinolysis.22
assess both bleeding and thrombosis risk in patients with The Multiplate Analyzer, first named the Whole-Blood Multiple
hepatic disease, those undergoing cardiac surgery, obstetric Electrode Platelet Aggregometer (MEA; Dynabyte, Munich,
patients, and trauma patients. Computerization of TEG facili- Germany, distributed by DiaPharma Group, West Chester,
tates its usefulness in intraoperative hemostasis management, Ohio), monitors the therapeutic efficacy of aspirin, clopido-
where it helps to predict the need for and monitor clotting grel, and glycoprotein IIb/IIIa antagonists using arachidonic
factor administration, platelet transfusion, fibrinolytic therapy, acid, ADP, and TRAP, respectively. The Multiplate provides mul-
and antiplatelet therapy with medications such as aspirin tiple channels with multiple electrodes per channel for simul-
or clopidogrel. Citrated whole blood is pipetted into a cylin- taneous analyses. In a representative study, Multiplate results
drical cup that oscillates by 4 degrees 45 minutes at a for specimens drawn before and after clopidogrel treatment
CHAPTER 47 Coagulation Instrumentation 791

central laboratory assays may be weak. Care must be taken to


Coagulation Fibrinolysis understand the differences between point-of-care and plasma-
based results and to ensure that dosing decisions based on the
LY point-of-care testing values are consistent. Table 47-3 summa-
Angle rizes the variety of FDA-approved point-of-care instruments
available.25
R K Ma

SELECTION OF
PT PFA-100 D-Dimer
COAGULATION INSTRUMENTATION
APTT Plateletworks
ACT OPA
In todays laboratory, more than ever before, cost effectiveness,
VerifyNow testing capabilities, and standardization are top priorities. As
an increasing number of tests become available, laboratories
Figure 47-6 Thromboelastograph (TEG) tracing from the TEG 5000 must determine what tests to incorporate to provide guidance
Hemostasis Analyzer System. The TEG analyzer produces results that docu to physicians in diagnosis and treatment. Identification of
ment the interaction of platelets with the protein coagulation cascade. The testing needs based on patient population should be the first
R value reflects initial fibrin clot formation (measured by the prothrombin time step in the process. The decisions regarding which tests are the
[PT], activated partial thromboplastin time [APTT], and activated clotting time most appropriate for the clinical situations encountered by
[ACT] assays). The K value shows the clot formation rate or the speed of the each laboratory should be made in conjunction with the
fibrin clot linking. The Ma value is an indicator of fibrin-platelet bonding medical staff. When that input has been obtained, the labora-
via glycoprotein IIb/IIIa (measured by the PFA-100, Plateletworks [Helena tory can determine the availability and cost of instruments that
Point of Care, Beaumont, Texas.], optical platelet aggregometry [OPA], would meet those requirements.
and VerifyNow). The LY shows the eventual clot lysis (measured by the
Instrument selection criteria may include, but are not
D-dimer assay). (The image of a hemostasis tracing from the TEG Hemo
limited to, the following:
stasis Analyzer is used by permission of the Haemoscope Corporation.)
Instrument cost
Consumables cost
Service response time
were compared with results obtained using a light-transmittance Reliability and downtime
aggregometer, the PAP-8E (Bio/Data Corporation, Horsham, Maintenance requirements and time
Pa.). The ADP-induced aggregation as measured by both instru- Operator ease of use
ments decreased significantly after clopidogrel administration, Breadth of testing menu
and the decreases measured by the two methods achieved Ability to add new testing protocols
a Spearman rank correlation coefficient of .71 (P < .0001). Throughput for high-volume testing
The group stated that the MEA was a fast and standardized Laboratory information systems interface capabilities
method of assessing platelet response to ADP before and after Footprint (the space the instrument occupies on the bench
clopidogrel treatment and that its results correlated well with top)
those of light transmission aggregometry. As of August 2010, Special requirements (water, power, waste drain)
Multiplate data were being prepared to obtain U.S. FDA Flexibility in using other manufacturers reagents
clearance.23,24 Availability of a training program and continued training
support
The laboratory director, manager, and technical staff may
POINT-OF-CARE TESTING
develop a comparison matrix listing each desired capability.
Point-of-care (bedside, near-patient) testing represents a When the choices have been narrowed based on the most desir-
growing segment of coagulation testing because of the need able criteria, consideration should be given to additional fea-
to monitor anticoagulant therapy. Point-of-care testing has tures. Because no instrument has all the features, prioritizing
enhanced the ability of the patient and physician to assess and helps the laboratory focus on the capabilities that would be the
modify anticoagulant dosage without the delays inherent most advantageous for them. Box 47-2 summarizes several of
in conventional testing and sometimes promotes patient these specialized features.
self-testing. An instrument should be matched to the anticipated work-
Point-of-care coagulation analyzers employ capillary or load. It may not be necessary to purchase a highly sophisticated
anticoagulated whole blood. For some instruments, a 10- to analyzer capable of performing a large menu of tests if the
50-mcL sample is transferred to a test cartridge and the cartridge setting is a small hospital laboratory ordering very few of the
is inserted into the test module. Other instruments require more esoteric test options available on the instrument under
500mcL to 2mL in a cuvette. Depending on the technology, consideration. A batch analyzer with high throughput may be
intra-assay correlation and correlation with plasma-based more appropriate for this situation.
TABLE 47-3 Review of Selected Point-of-Care Coagulometers
Manufacturer/ Specimen
Distributor Instrument Requirements Tests Performed Special Features
Abbott Point of Care, i-STAT 1 Capillary: fresh whole blood PT/INR, ACT (celite), ACT CHEM8+ (blood chemistry/electrolyte
Inc., Princeton, N.J. Venipuncture: citrated blood (kaolin) analysis, hematocrit, hemoglobin);
BNP, CK-MB, troponin 1 testing.
QC and data management
program with interface.
Helena Point of Care, Cascade POC Actalyke XL, Capillary: fresh whole blood PT/INR, celite ACT, Most of the special tests are
Beaumont, Tex. Actalyke Mini II Venipuncture: citrated blood low-molecular-weight performed on Cascade POC. Data
heparin MAX-ACT management for heparin dosing.
(reagent tube QC and data management.
supporting maximum
factor XII activation)
HemoSense/Inverness INRatio PT INR monitor, Capillary: fresh whole blood PT/INR Optional add-on CoaguClinic from
Medical, San Diego, INRatio2 PT INR monitor Standing Stone program,
Calif. interface to LIS.
Instrumentation Gem PCL Plus (portable Capillary: fresh whole blood PT/INR, ACT QC and data management to
Laboratory, coagulation laboratory) Venipuncture: citrated blood interface with LIS.
Bedford, Mass.
International ProTime Microcoagulation Capillary: ProTime systems PT/INR on ProTime QC and data management, program
Technidyne, Corp., System, ProTime Micro, Venipuncture: citrated systems, PT/INR, PT interface to LIS.
Edison, N.J. ProTime 3, New ProTime, blood, all systems citrate, ACT + ACT-LR,
Hemochron Signature HITT, TT, fibrinogen on
Elite, Signature Plus, Hemochron systems
Hemochron Response
Medtronic Cardiac HMS Plus, ACT Plus Venipuncture: citrated blood ACT, heparin dose Automated sample dispensing,
Surgery, response and multiple testing capability, data
Minneapolis, Minn. protamine titration management program, interface
to LIS.
Roche Diagnostics, CoaguChek XS and XS Plus Capillary: fresh whole blood PT/INR Data-management capability
Indianapolis, Ind. PT test system Venipuncture: citrated blood includes QC that interfaces to LIS.

ACT, Activated clotting time; ACT-LR, activated clotting time with low-range heparin plasma concentrations; BNP, brain natriuretic peptide; CK-MB, creatine kinase MB isoenzyme;
HITT, heparin-induced thrombocytopenia-thrombosis; INR, international normalized ratio; LIS, laboratory information system; POC, point of care; PT, prothrombin time; QC, quality
control; TT, thrombin time.

BOX 47-2 Specialized Coagulometer Features


Random access: A variety of tests can be performed on a single specimen Stat capabilities: Operator can interrupt a testing sequence to place a stat
or multiple specimens in any order as determined by the operator. specimen next in line for testing.
Primary tube sampling: Plasma is directly aspirated from an open or On-board refrigeration of specimens and reagents: Refrigeration maintains
capped centrifuged primary collection tube on the analyzer. the integrity of specimens and reagents throughout testing and allows
Cap piercing: Analyzer aspirates plasma from the closed centrifuged reagents to be kept in the analyzer for extended periods, which reduces
primary collection tube. setup time for less frequently performed tests.
Bar coding: Reagents and specimens are identified with a bar code; elimi On-board specimen storage capacity: Indicates the number of specimens
nates manual information entry. that can be loaded at a time.
Bidirectional laboratory information system (LIS) interface: Analyzer queries Reflex testing: Instrument can be programmed to perform repeat or addi
the host computer (LIS) to determine which tests have been ordered. tional testing under operator-defined circumstances.
Results are returned to the LIS after verification. Patient data storage: Test results can be stored for future retrieval; clot
Specimen and instrument flagging: Automated alerts indicate problems formation graphs may be included.
with specimen integrity or instrument malfunction. Throughput: Number of tests that can be processed within a specified
Liquid level sensing: Operator is alerted when there is inadequate speci interval, usually the number of tests per hour; depends on test mix and
men or reagent volume. An alert is also given when the instrument methodologies.
fails to aspirate the required sample volume. Volume is verified each Total testing (dwell) time: Length of time from specimen placement in the
time a specimen or reagent is aspirated. analyzer until testing is completed; depends on the type and complexity
On-board quality control: Instrument stores and organizes quality control of the procedure.
data; may include application of Westgard rules for flagging out-of- Graph of clot formation: Operator can visualize clot formation.
range results; instrument may transmit quality control data to the LIS.
CHAPTER 47 Coagulation Instrumentation 793

TABLE 47-4 Selected Fully Automated Coagulometers Currently Being Manufactured with the Largest U.S. Market Shares
Instrument End-Point Detection
Manufacturer/Distributor Series Instrument Name Method Special Features
Siemens Healthcare BCS, Sysmex BCS-XP, Sysmex Photo-optical, chromogenic, Primary tube sampling, bar coding, specimen
Diagnostics, Inc., series CA-7000, CA-1500, immunologic quality flagging, graph of clot formation,
Deerfield, Ill. CA-560, CA-530 bidirectional interface
Tcoag US, Parsippany, N.J. AMAX Destiny AMAX Destiny Optical, Mechanical, photo-optical, Bar coding, bidirectional interface, primary
series AMAX Destiny MAX, chromogenic, immunologic tube sampling, reduced reagent/specimen
AMAX Destiny Plus volumes, liquid level sensing, reflex testing
Instrumentation Laboratory/ ACL series ACL TOP, ACL Elite Nephelometry, photo-optical, Primary tube sampling, bar coding, batch
Beckman Coulter, Inc., series, ACL Advance chromogenic, immunologic analysis, bidirectional interface, graph of
Brea, Calif. series clot formation
Diagnostica Stago, Inc., STA series STA Compact and Mechanical, chromogenic, Bar coding for reagents and specimens,
Parsippany, N.J. Compact CT, STA-R immunologic bidirectional interface, simultaneous
Evolution Expert performance of end-point detection
series, STA Satellite methods, reflex testing, autoprogramming
for performance of quality control,
user-defined program
American LAbor, Durham, CoaLab series CoaLab 1000 Photo-optical Electronic signatures available for each
N.C. assay run, visualization and printouts,
positive displacement pipetting for low
maintenance and high precision, ability to
handle turbid/colored samples

considered for a particular laboratory setting and in developing


CURRENTLY AVAILABLE INSTRUMENTS
an organized approach for their evaluation. Table 47-4 lists
A variety of coagulometers address the increasing demand several of the coagulation analyzers currently available, the
for test volume, random access testing, and test variety. All type of end-point detection offered, and selected specialized
analyzers perform routine testing quickly and efficiently. The features highlighted by the manufacturers in their product
challenge lies in determining which instruments should be information.26

SUMMARY
Advanced technology used in semiautomated and automated Each method of end-point detection has advantages and disad-
analyzers has greatly improved coagulation testing accuracy and vantages that must be recognized and understood to ensure the
precision. validity of test results.
End-point detection methodologies employed by modern coagula- Point-of-care whole-blood analyzers are available to measure
tion analyzers include mechanical, photo-optical, nephelometric, platelet function in a more efficient and timely manner than tradi-
chromogenic, and immunologic methods. tional methods.
Advances in end-point detection methodologies have greatly Coagulation testing has been incorporated into the arena of point-
expanded the testing capabilities available in the routine coagula- of-care testing primarily to enhance the patients and physicians
tion laboratory. ability to monitor oral anticoagulant therapy.
Markedly improved instrument precision and reduced reagent A systematic approach to the evaluation and selection of a new
volume requirements have led to substantial cost savings in coagulation analyzer should be developed and followed to deter-
coagulation testing. mine the best instrument for a specific laboratory setting.
Instrument manufacturers have incorporated many features that
have enhanced efficiency, safety, and diagnostic capabilities in Now that you have completed this chapter, go back and
hemostasis testing. read again the case study at the beginning and respond
Coagulation analyzer flagging alert functions warn the operator to the questions presented.
when sample or instrument conditions exist that might lead to
invalid test results so that appropriate actions can be taken to
ensure test accuracy.
794 PART VIII Hemostasis and Thrombosis

R E V I E W Q UESTIONS
1. The photo-optical method of end-point detection can be 6. When a sample has been flagged as being icteric by an
described as: automated coagulation analyzer, which method would be
a. Measurement of a color-producing chromophore at a most susceptible to erroneous results because of the inter-
wavelength of 405nm fering substance?
b. Measurement of the change in OD of a test solution a. Mechanical clot detection
as a result of fibrin formation b. Immunologic antigen-antibody reaction detection
c. Application of an electromagnetic field to the test c. Photo-optical clot detection
cuvette to detect the decreased motion of an iron ball d. Chromogenic end-point detection
within the cuvette
d. Measurement of the turbidity of a test solution 7. Platelet function testing has been incorporated into
resulting from the formation of antigen-antibody the routine coagulation laboratory in recent years as a
complexes using latex particles result of:
a. Increased use of drugs that stimulate platelet
2. Modern coagulation analyzers have greatly enhanced the production in patients receiving chemotherapy
ability to perform coagulation testing as a result of which b. The convenience of being able to do the testing on
of the following? the same instrument that performs the coagulation
a. Maintenance of a level of accuracy and precision testing
similar to that of manual methods c. Increased therapeutic use of aspirin in the treatment
b. Increase in reagent volume capabilities to improve of heart disease
sensitivity d. Increased outpatient/outreach testing that prevents
c. Automatic adjustment of results for interfering the laboratory from having access to patients to do
substances bleeding time tests
d. Improved flagging capabilities to identify
problems in sample quality or instrument 8. All of the following are performance characteristics to con-
function sider in the selection of a coagulation analyzer except:
a. Location of the manufacturers home office
3. Which of the following is considered to be an advantage b. Instrument footprint
of the mechanical end-point detection methodology? c. Ease of use for the operator
a. It is not affected by lipemia in the test sample. d. Variety of tests the instrument can perform
b. It has the ability to provide a graph of clot
formation. 9. The PFA-100 measures platelet function by:
c. It can incorporate multiple wavelengths into a single a. Detecting the change in blood flow pressure along a
testing sequence. small tube when a clot impairs blood flow
d. It can measure proteins that do not have fibrin b. Detecting the aggregation of latex beads coated with
formation as the end point. platelet activators
c. Graphing the transmittance of light through platelet-
4. Which of the following methods use the principle of rich plasma over time after addition of platelet
changes in light scatter or transmission to detect the end activators
point of the reaction? d. Detecting the time it takes for a clot to form as blood
a. Immunologic, mechanical, photo-optical flows through a small aperture in a tube coated with
b. Photo-optical, nephelometric, mechanical platelet activators
c. Photo-optical, nephelometric, immunologic
d. Chromogenic, immunologic, mechanical 10. Point-of-care coagulation testing is used mainly:
a. To monitor patients receiving oral anticoagulant
5. Which of the following is a feature of semiautomated therapy
coagulation testing analyzers? b. To monitor patients taking platelet inhibitors such as
a. The temperature is maintained externally by a heat aspirin
block or water bath. c. To provide a baseline for all subsequent patient test
b. Reagents and samples usually are added manually result comparisons when the patient starts any kind
by the operator. of anticoagulant therapy
c. Timers are automatically started as soon d. To monitor obstetric patients at risk of fetal loss
as the analyzer adds reagents to the test
cuvette.
d. The end point must be detected by the
operator.
CHAPTER 47 Coagulation Instrumentation 795

REFERENCES
1. McGlinchey K: Sophistication in coagulation platforms. Adv 15. Harrison P, Mumford A: Screening tests of platelet function:
Med Lab Prof 20:20-21, 2009. Update on their appropriate uses for diagnostic testing. Semin
2. Owen CA: History of blood coagulation. JAMA 87:1051-1052, Thromb Hemost 35:150-157, 2009.
2002. 16. Franchini M: The platelet function analyzer (PFA-100): an
3. Bender GT: Principles of chemical instrumentation: STA-R update on its clinical use. Clin Lab 51:367-372, 2005.
operators manual, Parsippany, NJ, 1998, Diagnostica Stago, 17. Favaloro EJ: Clinical utility of the PFA-100. Semin Thromb
pp 119-124. Hemost 34:709-733, 2008.
4. Johns CS: Coagulation instrumentation. In Rodak BG, 18. McKee SA, Sane DC, Deliargyris EN: Aspirin resistance in
Fritsma GA, Doig K, editors: Hematology: clinical principles cardiovascular disease: A review of prevalence, mechanisms,
and applications, ed 3, St Louis, 2007, Saunders, pp 714- and clinical significance. Thromb Haemost 88:711-715, 2002.
728. 19. Lordkipanidze M, Pharand C, Schampaert E, et al: A compari-
5. Thomas LC, Sochynsky CL: Multiple measuring modes of son of six major platelet function tests to determine the
coagulation instruments. Clin Hemost Rev 13:8, 1999. prevalence of aspirin resistance in patients with stable coro-
6. Schueter A, Olson JD: Coagulation instrumentation: the nary artery disease. Eur Heart J 28:1702-1708, 2007.
hospital laboratory to the home. Adv Lab Methods Haematol 20. Dyskiewicz-Korpanty DM, Kim A, Durner JD, et al: Compari-
316-336, 2002. son of a rapid platelet function assayVerifyNow Aspirin
7. DiaPharma factor X kit. West Chester, Ohio, 2005, DiaPharma for the detection of aspirin resistance. Thromb Res 120:485-488,
Group (package insert). 2007.
8. STA Liatest D-Di. Parsippany, NJ, 2005, Diagnostica Stago 21. McGlasson DL, Fritsma GA: Comparison of four laboratory
(package insert). methods to assess aspirin sensitivity. Blood Coagul Fibrinolysis
9. Allen R, Sheridan B: Service above all. CAP Today, pp 39-60, 19:120-123, 2008.
January 1999. 22. Hobson AR, Agarwala RA, Swallow KD, et al: Thromboelas-
10. Kundu SK, Heilmann EF, Sio R, et al: Characterization of an tography: Current clinical applications and its potential role
in vitro platelet function analyzer, PFA-100. Clin Appl Thromb in interventional cardiology. Platelets 17:509-518, 2006.
Hemost 2:241-249, 1996. 23. Sibbing D, Braun S, Jawansky S, et al: Assessment of
11. Rodgers RPC, Levin J: A critical reappraisal of the bleeding ADP-induced platelet aggregation with light transmission
time. Semin Thromb Hemost 16:1-20, 1990. aggregometry and multiple electrode platelet aggregometry
12. Lind DE: The bleeding time does not predict surgical bleed- before and after clopidogrel treatment. Thromb Haemost 99:
ing. Blood 77:2547-2552, 1991. 121-126, 2008.
13. Gerwitz AS, Miller ML, Keys TF: The clinical usefulness of the 24. McGlasson DL, Fritsma GA: Whole blood aggregometry and
preoperative bleeding time. Arch Pathol Lab Med 120:353- platelet functional testing. Semin Thromb Hemost 35:168-180,
356, 1996. 2009.
14. Peterson P, Hayes TC, Arkin CF, et al: The preoperative bleed- 25. Coagulation analyzerspoint of care, self monitoring. CAP
ing time test lacks clinical benefit: College of American Today, pp 26-33, May 2009.
Pathologists and American Society of Clinical Pathologists 26. Dabkowski B: Coagulation analyzers offering speed, full
position article. Arch Surg 133:134-139, 1998. menus and more. CAP Today, pp 19-29, January 2009.
APPENDIX

Material Safety Data Sheet

Page 1 of 5
Material Safety Data Sheet
acc. to ISO/DIS 11014
Printing Date: 2010-Jan-14 Printing Date: Reviewed on
Product Name: BD Vacutainer Brand Tube with Dipotassium EDTA

1 Identification of substance:
Product Details:
MSDS Number: VS60311-18
Issue Date: January 14, 2010
Supersedes: VS60311-17
ECO No.: ECO118043
Product Name: BD Vacutainer Brand Tube with Dipotassium EDTA
Catalog Numbers: 363706, 365955, 365973, 365974, 365975, 366660, 366643, 366662, 367525, 367835,
367838 367840, 367841, 367842, 367843, 367844, 367845, 367846, 367855, 367856,
367859, 367861, 367862, 367863, 367864, 367899, 367950, 368021, 368047, 368048,
368049, 368054, 368171, 368267, 368381, 368589, 368661, 368841, 368861,
Discontinued: 360001, 360002, 360003, 360004, 367648, 367649, 367656, 367865,
367906, 367907, 367908, 367909
Manufacturer/Supplier:
BD Diagnostics, Preanalytical Systems
1 Becton Drive
Franklin Lakes, NJ, 07417-1885
Information Department:
BD Diagnostics, Preanalytical Systems Technical Service, (800) 631-0174
Emergency Information:
In case of a chemical emergency, spill, fire, exposure, or accident contact ChemTrec at (800) 424-9300

2 Composition/Data on components:
Description:
Chemical name: Dipotassium EDTA
CAS No.: 25102-12-9
Quantity: 1.0 29.0 mg
Exposure limits: N/A

3 Hazards identification:
Hazard description:
May cause eye and skin irritation. Causes respiratory tract irritation and gastrointestinal irritation. Avoid
generating dust or dust accumulation.
Acute Exposure Effects:
May cause skin and eye irritation. Inhalation causes respiratory tract irritation. Ingestion causes
gastrointestinal irritation with nausea, vomiting and diarrhea.
NFPA ratings (scale 0-4):
1 Health = 1
1 0 Fire = 1
Reactivity = 0
0
Specific Hazard =0

796
APPENDIX Material Safety Data Sheet 797

Page 2 of 5
Material Safety Data Sheet
acc. to ISO/DIS 11014
Printing Date: 2010-Jan-14 Printing Date: Reviewed on
Product Name: BD Vacutainer! Brand Tube with Dipotassium EDTA

HMIS ratings (scale 0-4):


1 Health = 1
1 0 Flammability = 1
0 Reactivity = 0
Special = 0

4 First aid measures:


General information:.
After inhalation: Move the exposed person to fresh air.
After skin contact: Immediately flush skin with plenty of soap and water for at least 15 minutes.
After eye contact: Immediately flush with plenty of water for at least 15 minutes.
After Swallowing: Immediately give large amounts of water. Get medical attention.
Information for doctor: Show this label

5 Fire fighting measures:


Suitable extinguishing agents: Use extinguishing media appropriate for surrounding fire.

Protective equipment: Firefighters should wear proper protective equipment and self-contained breathing
apparatus with full facepiece operated in positive pressure mode.

6 Accidental release measures:


Person-related safety precautions: Avoid overexposure. Wear suitable protective clothing.
Measures for environmental protection: N/A
Measures for cleaning/collecting: Carefully sweep up and remove. Flush area with water.
Additional information: N/A

7 Handling and storage:


Handling:
Information for safe handling: Keep container tightly closed. Suitable for any general chemical storage
area.
Information about protection against explosions and fires: Avoid contact with incompatible material,
minimize dust generation and accumulation.
Storage:
Requirements to be met by storerooms and receptacles: Keep container tightly closed.
Information about storage in one common storage facility: Suitable for any general chemical storage
area.
798 APPENDIX Material Safety Data Sheet

Page 3 of 5
Material Safety Data Sheet
acc. to ISO/DIS 11014
Printing Date: 2010-Jan-14 Printing Date: Reviewed on
Product Name: BD Vacutainer! Brand Tube with Dipotassium EDTA

Further information about storage conditions: None

8 Exposure controls and personal protection:


Additional information about design of technical systems: No further data
Components with limit values that require monitoring at the workplace: N/A
Additional information: N/A
Personal Protective Equipment:
General protective and hygienic measures: Wash thoroughly after handling. Remove contaminated
clothing and wash before reuse. Avoid contact with eyes, skin, and clothing. Avoid ingestion and
inhalation. Use with adequate ventilation.
Breathing equipment: None required where adequate ventilation conditions exist. If airborne
concentration is high, use an appropriate dust mask.
Protection of hands: Use proper gloves
Eye protection: Use safety goggles
Body protection: Wear appropriate protective clothing to prevent skin exposure.

09 Physical and chemical properties:


General Information:
Form: Solid (Crystalline powder)
Color: White
Odor: Odorless
Change in condition:
Melting point/Melting range: Not available
Boiling point/Boiling range: Not available
Flash point: N/A
Flammability (solid, gaseous): N/A
Danger of explosion: This material may cause a dust explosion
Vapor pressure: N/A
Density: N/A
Solubility in/Miscibility w/H2O: 100%
pH-value: Not available
Organic solvents: Not available
Solids content: Not available
APPENDIX Material Safety Data Sheet 799

Page 4 of 5
Material Safety Data Sheet
acc. to ISO/DIS 11014
Printing Date: 2010-Jan-14 Printing Date: Reviewed on
Product Name: BD Vacutainer! Brand Tube with Dipotassium EDTA

10 Stability and reactivity:


Thermal decomposition / conditions to be avoided: Avoid dust generation
Materials to be avoided: None identified
Dangerous reactions: No dangerous reactions known.
Dangerous products of decomposition: Oxides of nitrogen, carbon monoxide, carbon dioxide

11 Toxicological information:
Acute toxicity:
Eye: May cause eye irritation
Skin: May cause skin irritation
Inhalation: Causes respiratory tract irritation
Ingestion: Causes digestive tract irritation
Primary irritant effect:
On the skin: Not established
On the eye: Not established
Sensitization: Not established
Additional toxicological information: Does not contain more than 0.1% by weight of a material listed as
a potential carcinogen by NTP, IARC, or OSHA.

12 Ecological information:
Ecotoxicological effects: No data is available on the adverse effects of this material on the environment.
Neither COD nor BOD data are available.
Other information: N/A
General notes: N/A

13 Disposal considerations:
Product:
Recommendation
Disposal should be done in accordance with local, state and federal regulations.
Disposal must be made according to the regulations found in 40 CFR 261.
This product is not a RCRA hazardous waste.
Uncleaned packagings:
Recommendation
Disposal should be done in accordance with local, state and federal regulations.
Disposal must be made according to the regulations found in 40 CFR 261.
This product is not a RCRA hazardous waste.
Recommended cleansing agent
Water, if necessary with cleansing agents
General notes: N/A
800 APPENDIX Material Safety Data Sheet

Page 5 of 5
Material Safety Data Sheet
acc. to ISO/DIS 11014
Printing Date: 2010-Jan-14 Printing Date: Reviewed on
Product Name: BD Vacutainer! Brand Tube with Dipotassium EDTA

14 Transport information:
DOT regulations: Non-regulated

Land transport ADR/RID (cross-border): N/A

Maritime transport IMDG: Non-regulated

Air transport ICAO-TI and IATA-DGR: Non-regulated

15 Regulations:
SARA Section 355 (extremely hazardous substances): Substance not listed
Section 313 (specific toxic chemical listings): None
TSCA (Toxic Substances Control Act) Inventory: Listed
California Proposition 65 Chemicals Known to Cause Cancer: Substance not listed
California Proposition 65 Chemicals Known to Cause Reproductive Toxicity: Substance not listed
Carncinogenicity categories
IARC (International Agency for Research on Cancer): Substance not listed
NTP (National Toxicology Program): Substance not listed
TLV (Threshold Limit Value established by ACGIH): Substance not listed
MAK (German Maximum Workplace Concentration): Not available
Product related hazard information: Minimize dust generation or accumulation
National regulations: N/A
Water hazard class: N/A

16 Other information:
To the best of our knowledge, the information contained herein is accurate. However, neither BD or any of
its subsidiaries assumes any liabilities whatsoever for the accuracy or completeness of the information
contained herein. Final determination of suitability of any material is the sole responsibility of the user. All
materials may present unknown hazards and should be used with caution. Although certain hazards are
described herein, we cannot guarantee that these are the only hazards that exist.
Department issuing MSDS: Regulatory Affairs

Courtesy and Becton, Dickinson, and Company.


Answers

3. c
CHAPTER 2
4. c
Case Study 5. a
1. This item describes two deficiencies. First, the hematology 6. b
technologist was not following the laboratory policy on 7. c
handwashing. Hands are washed after removing gloves 8. b
and before leaving the laboratory. Second, the hematology 9. b
technologist should have removed the laboratory coat 10. b
before going to the meeting. A second laboratory coat
should have been used if it was necessary to wear a labora-
CHAPTER 3
tory coat outside the laboratory.
2. This item describes a deficiency. Storage of food in a speci- Case Studies
men refrigerator is prohibited. Case 1
3. This item describes a deficiency. Needles should not be Proper procedure is to ask the patient to state his or her name
recapped unless there is a specific medical procedure that and confirm by asking the birth date or Social Security number.
requires recapping. No methods used in hematology Tubes are never prelabeled. They are labeled after blood is
justify recapping of the syringe. drawn and before leaving the patient.
4. This may or may not be a deficiency. The technologist may
have had on a personal laboratory coat. A second labora- Case 2
tory coat could have been obtained by the technologist to Results that can be affected by this order of draw include PT
wear in public areas. and K+.
5. No deficiency is indicated. Fire extinguishers should be The gel separator tube should not be used for blood for the
placed every 75 feet. blood bank because it contains clot activator. This specimen
6. No deficiency is indicated. Fire extinguishers should be should not be drawn before a specimen for coagulation testing,
checked monthly and annually. because carryover could contaminate the specimen. If K+ is
7. This item represents a deficiency. Quarterly fire drills being tested from blood in a green tube, the specimen must be
should be conducted. drawn before the EDTA tube is filled. EDTA is usually in a K+
8. This item represents a deficiency. All chemicals should be salt, which could falsely elevate K+ levels in blood drawn after
labeled. the EDTA tube is filled.
9. This item represents a deficiency. The 1:10 bleach solu- See correct order of draw for evacuated tubes on page 24
tions should be made fresh daily. Step 13.
10. No deficiency is indicated. Gloves should be worn by all
personnel handling specimens. Review Questions
11. No deficiency is indicated. Material safety data sheets can 1. c
be received electronically. 2. b
12. This item describes a deficiency. Chemicals should not be 3. b
stored under the hood and should not be stored 4. b
alphabetically. 5. b
13. This item describes a deficiency. All staff members should 6. d
participate in scheduled fire drills. 7. b
8. c
Review Questions 9. d
1. d 10. b
2. b 11. a

801
802 APPENDIX Answers

10. c
CHAPTER 4
11. a
Case Study 12. c
The following things should be checked:
Is the slide right side up? This is the most common cause
CHAPTER 6
of inability to focus a slide under oil when it has been focused
under 10 and 40 objectives. Review Questions
If the slide is right side up, continue by checking the 1. b
following: 2. c
Is there sufficient oil on the slide? If not, apply another drop 3. a
of oil. 4. b
Is the objective screwed in tightly? If not, tighten the 5. c
objective. 6. a
If the slide is coverslipped, is there more than one coverslip 7. d
on the slide? If so, gently remove the top coverslip. 8. d
Has oil seeped into the seal on the oil objective? Examine 9. c
by removing the objective and using an inverted ocular as 10. b
a magnifier to check the seal. If the seal is broken, the objec- 11. a
tive must be replaced. 12. d
13. b
Review Questions
1. b
CHAPTER 7
2. c
3. a Review Questions
4. b 1. a
5. c 2. d
6. d 3. c
7. a 4. a
8. c 5. b
9. d 6. c
10. b 7. b
8. b
9. a
CHAPTER 5
10. c
Case Study
1. This is a systematic error because the magnitude of error
CHAPTER 8
remains constant at three ranges of test results for two runs.
2. It is not acceptable to continue using the instrument or to Case Study
simply subtract the systematic error from test sample results. 1. When the blood is not well oxygenated, the bone marrow
All the samples in the two out-of-control test runs must be responds by producing more red blood cells to carry more
re-assayed after the error is corrected. oxygen.
3. Determine from the quality control charts at what moment 2. The hormone that stimulates red blood cell production is
the error occurred. Investigate potential changes in instru- erythropoietin. The peritubular cells of the kidney detect
ment settings, calibration, reagent changes, or instrument hypoxia. A hypoxia-sensitive transcription factor is pro-
malfunctions that may have occurred at the instant the error duced that moves to the nucleus and upregulates trans
is recorded. cription of the erythropoietin gene. Erythropoietin acts by
preventing apoptosis of the erythroid colony-forming unit.
Review Questions It stimulates production of antiapoptotic molecules.
1. c 3. Once the patient was receiving oxygen therapy, hypoxia
2. b diminished and erythropoietin production also declined.
3. b Thus, production of new red blood cells slowed. At the same
4. a time, red blood cells reaching 120 days of age died off. Thus
5. d the total number of red blood cells decreased.
6. d
7. c Review Questions
8. c 1. c
9. a 2. a
APPENDIX Answers 803

3. d 4. Infants have a high HbF level, which consists of and


4. b chains. As the chain gene becomes acetylated, chain
5. b production begins to predominate, producing HbA, adult
6. d hemoglobin. These are the expected results for Hb electro-
7. a phoresis for mother and infant.
8. d
9. a Review Questions
10. c 1. d
11. b 2. a
12. b 3. a
4. a
5. b
CHAPTER 9
6. d
Case Study 7. c
1. A reducing agent is able to donate an electron to an oxidized 8. b
compound so that the oxidized compound has one fewer 9. d
unpaired protons. The compound receiving the electron 10. b
becomes reduced and the donating compound becomes 11. c
oxidized.
2. When heme iron is oxidized, the molecule cannot carry
CHAPTER 11
oxygen and patients become cyanotic. Because vitamin C
eliminated the cyanosis, it must be able to reduce methemo- Case Study
globin and restore the oxygen-carrying capacity of the blood. 1. Iron loss via blood donations and obligatory loss was not
3. Because this condition affected brothers, a hereditary condi- compensated by diet or supplementation.
tion was suggested in which hemoglobin became oxidized 2. Average daily iron absorption rate in this age group is 1 to
more than is usual. (The condition affecting these brothers 2mg/d.
was later identified as a hereditary deficiency of methemo- 3. Adaptation: when storage iron declines, hepcidin levels
globin reductase.) decline, which induces increased duodenal absorption.
4. Serum iron, total iron-binding capacity, transferrin, trans-
Review Questions ferrin saturation, and ferritin. Additional tests are red blood
1. b cell protoporphyrin, serum transferrin receptor, and bone
2. a marrow iron stores.
3. a 5. See Table 11-2.
4. c 6. The serum ferritin test is the only one that estimates iron stores.
5. c
6. c Review Questions
7. d 1. c
8. a 2. b
9. c 3. d
10. b 4. b
11. d 5. c
12. b 6. b
13. a 7. d
8. c
9. b
CHAPTER 10
10. d
Case Study
1. Yes. The hemoglobin reference interval for adult women is
CHAPTER 12
12.0 to 15.0g/dL and for newborns is 16.5 to 21.5g/dL.
The mothers and infants results were within the range. Review Questions
2. Birth hemoglobin is high because HbF weakly binds 2,3- 1. b
BPG. The hemoglobin concentration compensates for its 2. d
high oxygen affinity and reduced oxygen transfer to tissues. 3. a
The hemoglobin concentration decreases to near adult 4. c
intervals at 1 year of age. 5. b
3. The hemoglobin assay measures concentration; electropho- 6. c
resis identifies hemoglobin types. 7. b
804 APPENDIX Answers

8. c Case 3
9. a 1. Sodium concentration. The sample electrolyte concentra-
10. b tion is used to correct the measured conductivity prior to
reporting hematocrit results. Factors that affect sodium con-
centration will therefore also affect the hematocrit.
CHAPTER 13
2. A high sodium concentration would falsely decrease the
Case Study hematocrit.
1. Bleeding characterized by petechiae, purpura, and ecchymo- 3. Factors that decrease the hematocrit are low total protein,
ses is known as mucocutaneous bleeding, also called systemic settling of red blood cells in the collection device, and con-
bleeding. By contrast, anatomic bleeding is bleeding into soft tamination by intravenous solutions.
tissue, muscles, joints, or body cavities.
2. Thrombocytopenia, or low platelet count, is a common cause Review Questions
of mucocutaneous bleeding. Another is diseases that weaken 1. b
vascular collagen such as scurvy. 2. c
3. No, the bone marrow megakaryocyte estimate is high, indi- 3. c
cating an increase in platelet production. 4. d
4. Thrombopoietin and interleukin-11 have the greatest effect on 5. d
recruitment and proliferation of megakaryocytes and their 6. b
progenitors. Also involved in early progenitor recruitment 7. c
are interleukin-3 and interleukin-6. Other cytokines and 8. c
hormones that participate synergistically with thrombo 9. a
poietin and the interleukins are stem cell factor, also called 10. d
kit ligand or mast cell growth factor; granulocyte-macrophage
colony-stimulating factor; granulocyte colony-stimulating
CHAPTER 15
factor; and erythropoietin.
Case Study
Review Questions The patients hemoglobin shows neither anemia or polycythe-
1. d mia; hence it is normal. Red blood cells are normocytic and
2. d normochromic with no anisocytosis. The blood picture shows
3. c leukocytosis and thrombocytopenia. The mean platelet volume
4. b is slightly low, which suggests small average platelet size. There
5. d is no white blood cell (WBC) differential.
6. d 1. The platelet count and WBC count should be questioned
7. a because of platelet clumping. EDTA-induced pseudothrom-
8. a bocytopenia and pseudoleukocytosis most likely occurred.
9. c 2. The specimen should be redrawn in sodium citrate and
10. d processed through the automated analyzer. The new WBC
and platelet counts should then be adjusted for the sodium
citrate dilution by multiplying the results by the dilution
CHAPTER 14
factor 10/9 or 1.1. The following are the new results:
Case Studies a. WBCs for specimen drawn in sodium citrate: (8.4
Case 1 109/L) 1.1 = 9.2 109/L (the corrected WBC count)
1. Hb 3 = Hct 3 b. Platelets for specimen drawn in sodium citrate: (231
15 3 = 45 3 (42-48) 109/L) 1.1 = 254 109/L (the corrected platelet
2. Lipemia, increased white blood cells (WBCs), abnormal count)
hemoglobin.
3. Lipemia: perform a plasma blank. Review Questions
Increased WBCs: centrifuge the hemoglobin solution and 1. d
read the transmittance of the supernatant. 2. c
Abnormal hemoglobin: add potassium carbonate to the 3. a
prepared dilution of blood and reagent. 4. c
5. b
Case 2 6. c
1. Mean cell volume = 59fL; mean cell hemoglobin = 18.1pg; 7. a
mean cell hemoglobin concentration = 30.7g/dL. 8. b
2. Microcytic, hypochromic red blood cells. 9. Leukocytosis with a relative neutrophilia (includes the
3. Examine the patients peripheral blood film. young neutrophilic cells). There is a dramatic absolute
APPENDIX Answers 805

neutrophilia with a left shift back to blasts but also an


CHAPTER 18
absolute lymphocytosis, eosinophilia, and basophilia. Red
blood cell morphology is not mentioned and thus is Case Study
normal. 1. Anemia is not a disease or diagnosis in itself but is the
10. There is a microcytic, hypochromic anemia with anisocy- symptom of an underlying disorder. A complete history and
tosis. The latter is confirmed in the morphology descrip- physical examination are necessary to help identify the
tion, which also notes poikilocytosis but does not describe cause(s) of the anemia. If the underlying cause is not
any particular cell shape. No mention of polychromasia or determined and corrected, the patient will continue to be
inclusions. anemic. Questions regarding lifestyle, medications, and
bleeding history are only some of the questions that should
be asked.
CHAPTER 16
2. The reticulocyte count differentiates anemias into those
Case Study involving impaired production (decreased reticulocyte
1. Bone marrow cellularity, estimated from the core biopsy count) and increased destruction (increased reticulocyte
specimen, or the aspirate if a biopsy specimen is unavail- count). If the reticulocyte is low, the anemia can be further
able, provides information on blood cell production. subdivided on the basis of mean cell volume into normo-
2. The ratio is 9:1, which indicates myeloid hyperplasia. cytic, microcytic, or macrocytic. With that knowledge,
3. When a bone marrow aspirate or core biopsy specimen is appropriate laboratory testing can be ordered to determine
reviewed, the normal megakaryocyte distribution is 2 to the cause.
10 per low-power field. Counts outside these limits are 3. The peripheral blood film yields valuable information
characterized as decreased or increased megakaryocytes. about the volume and hemoglobin content of the erythro-
Megakaryocyte morphology is also reviewed for diameter, cytes as well as any abnormal shapes, which may be corre-
granularity, and nuclear lobularity. lated with specific causes. Some anemias are also associated
with white blood cell and/or platelet abnormalities, which
Review Questions may be noted on the blood film.
1. c
2. b Review Questions
3. c 1. c
4. a 2. b
5. d 3. a
6. c 4. c
7. d 5. a
8. b 6. b
9. b 7. c
10. b 8. d
11. b 9. b
10. a

CHAPTER 17
CHAPTER 19
Case Study
1. Tube 3 or the least bloody tube. Case Study
2. A 1:53 dilution with saline is necessary for a satisfactory 1. Hypochromic, microcytic anemias to be considered include
cytocentrifuge slide. iron deficiency anemia, thalassemia, sideroblastic anemias,
3. Bacteria. and, possibly, anemia of chronic inflammation.
4. The most likely diagnosis is bacterial meningitis. 2. Thalassemia can be eliminated because it is not a condition
that would be acquired late in life. Although iron deficiency
Review Questions anemia is not as common in women after menopause, it is
1. b probably the most likely of the remaining possibilities for
2. a an anemia this severe.
3. c 3. Anemia of chronic inflammation could be eliminated in
4. b this case because the woman is otherwise healthy. Iron defi-
5. a ciency anemia is probably more likely.
6. b 4. Iron studies, including ferritin, would be useful in clarifying
7. c the patients diagnosis. Assuming that she is iron deficient,
8. d the ferritin, total serum iron, and percent saturation should
9. c all be decreased, whereas total iron-binding capacity (TIBC)
10. a would be expected to be increased. Upon hospitalization,
806 APPENDIX Answers

the patient was immediately placed on oxygen while labora- 3. The patients vitamin assays point to a deficiency of
tory tests were ordered. With the confirmation by vitamin B12, substantiated by an increase in methylmalonic
the hospital laboratory of a dangerously low hemoglobin, acid.
transfusions were ordered, and the patient received 3 units 4. Based on these results, tests for antibodies to parietal cells
of packed cells over the first 2 days of hospitalization. The and intrinsic factor would be appropriate. However, the
transfusions were administered very slowly so as not to physician also inquired further about the patients dietary
stress her cardiovascular system with added volume. Noting habits and learned that he enjoyed dishes of raw fish
the hypochromic, microcytic blood picture, the physician obtained from the surrounding lakes. Therefore, the physi-
ordered iron studies, for which specimens were drawn prior cian ordered a stool analysis for ova and parasites. The
to transfusion. The results were as follows: serum iron study indicated the presence in the stool of both eggs and
decreased, TIBC increased, percent saturation decreased, proglottids of the fish tapeworm Diphyllobothrium latum.
and ferritin decreased. The possibility of gastrointestinal The patient was treated with a suitable purgative, and the
bleeding as a cause for iron deficiency was investigated. scolex of the tapeworm was discovered in a stool sample
Results of tests for occult blood in the stool were negative. after a single treatment. The patient was counseled on the
The hospital dietitian assessed the patients usual diet of tea, proper preparation of fresh fish to avoid reinfection. He
toast, canned soup, and crackers and determined that it was received injections of cyanocobalamin to replenish his
quite inadequate not only in iron, but also in other impor- vitamin B12 stores. His hemoglobin returned to normal
tant nutrients. The physician concluded that the patients over the next month, and his neurologic symptoms
dietary iron deficiency had developed slowly, which had subsided.
allowed her to adapt to an exceedingly low hemoglobin
level. Furthermore, her low level of activity meant that she Review Questions
rarely experienced the effects of the anemia. She was started 1. d
on a course of oral iron supplementation, and arrange- 2. c
ments were made for her to receive one balanced meal daily 3. c
from the Meals on Wheels program sponsored through a 4. b
community service organization for senior citizens. She 5. a
was quite responsible about taking her iron supplements, 6. b
and her hemoglobin was within the reference range within 7. c
3 months. 8. d
9. a
Review Questions 10. c
1. b
2. a
CHAPTER 21
3. d
4. a Case Study
5. c 1. The term used to describe a decrease in all cell lines is
6. a pancytopenia.
7. d 2. Acquired aplastic anemia should be considered due to
8. c the pancytopenia, reticulocytopenia, bone marrow hypo
9. b cellularity, normal vitamin B12 and folate levels, absence of
10. d blasts and abnormal cells in the bone marrow and periph-
eral blood, normal myelopoiesis and megakaryopoiesis,
and history of a prescribed nonsteroidal antiinflammatory
CHAPTER 20
agent.
Case Study 3. An increase in blasts or reticulin in the bone marrow sug-
1. The complete blood count findings for this patient (notably gests a diagnosis of myelodysplasia or leukemia.
macrocytic, normochromic anemia; pancytopenia; hyper- 4. The extent of the patients bone marrow hypocellularity
segmentation of neutrophils; and oval macrocytes) were and her hemoglobin and neutrophil and platelet counts
consistent with the physicians suspicion of megaloblastic place her disorder in the nonsevere aplastic anemia
anemia as suggested by the clinical findings. category.
2. Although the relative reticulocyte count was slightly above 5. Due to the patients age and the fact that her disorder
the reference range of 0.5% to 1.5%, and the calculated is in the nonsevere category, immunosuppression with
absolute reticulocyte count (approximately 40 109/L) was antithymocyte globulin and cyclosporine is the preferred
within the reference range of 25 to 75 109/L, the calculated treatment. In general, red cells would be transfused if
reticulocyte production index was 0.5, which was clearly the patient had symptoms of anemia, whereas platelet
inadequate to compensate for a substantial anemia (see transfusions would be given if her platelet count fell below
Chapter 14). 10 109/L.
APPENDIX Answers 807
Review Questions bilirubin level and decreased serum haptoglobin level),
1. c increased erythropoiesis to compensate for the premature
2. d hemolysis (increased reticulocyte count), and the nonim-
3. b mune nature of the hemolysis (negative result on the direct
4. d antiglobulin test). Testing family members to establish a
5. b mode of inheritance is desirable. The osmotic fragility test
6. d is expected to show increased fragility, but that test and
7. c other special tests are not required for diagnosis of HS in a
8. d patient with a familial inheritance pattern and the typical
9. c clinical and laboratory findings (see Table 23-2).
10. d 3. HS is an inherited intrinsic hemolytic anemia caused by a
11. a mutation that disrupts the vertical protein interactions in the
red blood cell (RBC) membrane. Various mutations in six
known genes can result in the HS phenotype (see Table
CHAPTER 22
23-1). The defective membrane protein causes the RBCs to
Case Study lose unsupported lipid membrane over time due to a local
1. Intravascular hemolysis is suspected in this patient, because disconnection between transmembrane proteins and the
the color of the urine is likely due to oxidized hemoglobin. cytoskeleton. The loss of membrane with minimal loss of cell
2. Tests for serum haptoglobin, serum indirect bilirubin, volume results in a decreased surface areatovolume ratio
serum lactate dehydrogenase, free plasma hemoglobin, and and the formation of spherocytes. Spherocytes do not have
urine hemoglobin, and examination of a peripheral blood the deformability of normal biconcave discoid RBCs. As the
film can differentiate the cause of hemolysis as intravascular cells repeatedly go through the spleen, they lose more mem-
or extravascular. brane due to splenic conditioning and eventually become
3. Due to the likelihood that the patient had hemoglobinuria, trapped in the spleen and removed by the splenic macro-
intravascular hemolysis is suspected. Therefore, the serum phages. The RBC membrane also has abnormal permeability
haptoglobin would be markedly decreased and the serum to cations, particularly sodium and potassium, likely due to
lactate dehydrogenase and free plasma hemoglobin would the disruption of the cytoskeleton by the mutated protein.
be increased. Routine urinalysis should yield positive results
for blood with no intact red blood cells in the urine sedi- Review Questions
ment. The serum bilirubin does not increase immediately 1. b
after an episode of intravascular hemolysis, but should 2. a
begin to increase within several days. The peripheral blood 3. a
film may demonstrate schistocytes. 4. b
5. a
Review Questions 6. d
1. a 7. a
2. b 8. b
3. d 9. b
4. b 10. a
5. c 11. c
6. a
7. d
CHAPTER 24
8. b
9. c Case Study
10. c 1. Many malarial ring forms, with multiple ring forms in indi-
vidual red blood cells (RBCs), are present in the thin
peripheral blood film. Many ring forms are also present in
CHAPTER 23
the thick peripheral blood film.
Case Study 2. The high parasitemia, the presence of multiple ring forms
1. On the basis of the patients jaundice and splenomegaly, in individual RBCs, and the absence of other parasite stages
history of gallstones, family history of anemia, low hemo- in the thin and thick peripheral blood films suggest a diag-
globin, increased mean cell hemoglobin concentration and nosis of malaria due to Plasmodium falciparum.
red cell distribution width, and spherocytes and polychro- 3. The patient had typical symptoms of malaria after a recent
masia on the peripheral blood film, hereditary spherocyto- 3-week trip to Ghana in West Africa. Malaria is endemic in
sis (HS) is suspected. Ghana, and according to the Centers for Disease Control
2. Additional laboratory tests to confirm HS should demon- and Prevention,57 85% of the malaria cases in Ghana are
strate increased hemolysis (increased serum indirect due to P. falciparum.
808 APPENDIX Answers

4. The only other forms of P. falciparum that are seen on a adsorption mechanism. When the drug adsorption mecha-
peripheral blood film are gametocytes, and they are charac- nism is causing the hemolysis, patient serum will react only
teristically crescent shaped. with drug-treated panel cells.
5. Anemia in malaria is due to (1) direct lysis of infected RBCs 5. The drug should be discontinued immediately.
during schizogony; (2) immune destruction of infected and
noninfected RBCs due to the binding of malarial proteins Review Questions
(shed during the invasion process) to the membranes of 1. a
infected and uninfected RBCs, subsequent binding of anti- 2. a
bodies and complement to the damaged RBCs, and clear- 3. d
ance of these RBCs by the macrophages in the spleen; and 4. c
(3) inhibition of erythropoiesis. 5. c
6. b
Review Questions 7. c
1. c 8. c
2. a 9. c
3. b 10. c
4. b
5. c
CHAPTER 26
6. b
7. c Case Study
8. c 1. The confirmatory test that should be performed is citrate agar
9. c electrophoresis at a pH between 6.0 and 6.2. In the citrate
10. d agar test, Hb C is separated from Hb A2, Hb O, and Hb E,
11. c and Hb S is separated from Hb D and Hb G (see Figure 26-7).
2. The characteristic morphologic feature on the peripheral
blood film is Hb SC crystals. They appear as fingerlike or
CHAPTER 25
quartzlike crystals of dense hemoglobin protruding from
Case Study the RBC membrane.
1. Because the patient was previously healthy, the anemia in 3. On the basis of the electrophoretic pattern and RBC mor-
this patient is most likely an acquired condition and extrin- phology, Hb SC disease is likely.
sic to the red blood cells (RBCs). 4. With parents of the genotypes SC and AS, 25% of the off-
2. A positive result on the direct antiglobulin test (DAT) is due spring would have each of the following genotypes: AS, SS,
to the binding of an immunoglobulin G (IgG) antibody, AC, and SC.
C3d, or both to the RBC surface and is evidence of an
immune cause of the hemolytic anemia. Review Questions
3. Three mechanisms that cause drug-induced immune hemo- 1. d
lytic anemia are the following: (1) drug adsorbs to the RBC 2. b
surface, the patients antidrug antibody (IgG) binds to the 3. c
drug, and the spleen then clears the antibody- and drug- 4. b
coated RBCs from circulation (extravascular hemolysis); 5. d
(2) an immunogenic complex is formed by drug bound 6. b
loosely to an RBC membrane protein, and the patients IgG or 7. a
IgM antibody binds to the complex, activates complement, 8. b
and causes acute intravascular hemolysis; (3) the drug induces 9. c
the production of IgG, warm-reacting RBC autoantibodies. 10. a
4. The mechanism most probably causing the patients anemia
is the drug adsorption mechanism. The drug is strongly
CHAPTER 27
bound to the patients RBCs. The patient produces an anti-
drug antibody (IgG class) that binds to the drug on the Case Study
surface of the RBC, which causes the positive DAT result. 1. The family history revealed a Mediterranean ethnic back-
The presence of spherocytes on the peripheral blood film ground; both - and -thalassemia are common in the
and the absence of hemoglobinuria indicate that the hemo- Mediterranean population. The students mother had always
lysis is likely extravascular. The negative results for the indi- been anemic, and her gallbladder attacks were probably
rect antiglobulin test on all panel cells tested with the caused by pigmented gallstones (calcium bilirubinate),
patients serum and an eluate prepared from the patients which resulted from the mild hemolytic anemia of hetero-
RBCs is evidence against induction of an RBC autoantibody, zygous -thalassemia. A cousin on the mothers side had
and is the typical finding in anemia due to the drug children with thalassemia major. Because of the family
APPENDIX Answers 809

history, it is quite likely that the student has -thalassemia 3. No. Some organisms make hydrogen peroxide and can
minor. Note that his mother was periodically given iron therefore be killed.
therapy. It is a common mistake to treat a thalassemic indi- 4. The majority of cases are X-linked.
vidual for iron deficiency anemia, especially in areas in
which thalassemia is not common in the general popula- Case 2
tion, because both iron deficiency anemia and thalassemia 1. Because of the reactive monocytosis, the blood film should
are microcytic, hypochromic anemias. be examined for possible circulating macrophages.
2. The student had a mild hypochromic (decreased mean 2. On the edges of the blood film, because macrophages are
cell hemoglobin concentration) and microcytic (decreased very large cells.
mean cell volume) anemia with target cells and basophilic 3. Circulating macrophages indicate sepsis.
stippling on his peripheral blood film. He had an elevated 4. A buffy coat preparation, which concentrates nucleated
level of hemoglobin A2, which is a marker for -thalassemia cells.
minor. His serum ferritin level was within the reference
range, which ruled out a diagnosis of iron deficiency anemia. Review Questions
3. A microcytic, hypochromic anemia could be due to - or 1. a
-thalassemia, Hb E disease or trait, iron deficiency anemia, 2. d
or, more rarely, sideroblastic anemia (including lead poi- 3. c
soning), or anemia of chronic inflammation (see Figure 4. b
18-3). Iron deficiency anemia is the most common of these. 5. d
Iron studies can differentiate these conditions (see Table 6. c
19-1). An incorrect presumption that a patient has iron 7. d
deficiency may lead to inappropriate iron therapy or to 8. c
unnecessary diagnostic procedures. 9. b
4. The potential mother should be screened for -thalassemia 10. a
trait, and if she is heterozygous for a -thalassemia gene
mutation, the couple should be advised that there is a 25%
CHAPTER 29
chance of having a baby with -thalassemia major (homo-
zygous or compound heterozygous for a -thalassemia Review Questions
mutation). In addition, there is a 25% chance of having 1. b
a baby who is homozygous for normal -globin genes, 2. b
and a 50% chance of having a baby heterozygous for a 3. c
-thalassemia mutation (-thalassemia trait). 4. c
5. a
Review Questions 6. d
1. b 7. d
2. c 8. b
3. a 9. c
4. d 10. a
5. a
6. c
CHAPTER 30
7. a
8. c Case Study
9. d 1. -Naphthyl acetate and butyrate esterases stain cells of
10. a monocytic origin. Chloroacetate esterase stains neutrophils.
11. d 2. The general diagnosis suggested by the elevated white
12. b blood cell count and decreased hemoglobin and platelet
13. c count, along with the presence of immature cells, is acute
14. d leukemia.
15. c 3. The most likely diagnosis is acute monocytic leukemia.

Review Questions
CHAPTER 28
1. b
Case Studies 2. c
Case 1 3. b
1. Chronic granulomatous disease. 4. d
2. Patient neutrophils are unable to form reactive oxygen 5. d
species such as hydrogen peroxide. 6. b
810 APPENDIX Answers

7. b 4. The following band sizes are expected in factor V DNA


8. c analysis:
9. c Normal: 104 and 82bp (37bp is sometimes barely visible,
10. a as well)
Heterozygous: 141, 104, and 82bp (37bp is sometimes
barely visible, as well)
CHAPTER 31
Homozygous: 141 and 82bp
Case Study 5. The initial blood specimen drawn from this patient was in
1. G banding utilizes Giemsa staining to differentiate chromo- a red-topped tube for the recovery of serum. Because red-
somes into bands for identification of specific chromo- topped tubes do not preserve the white blood cells needed
somes. The chromosomes must be pretreated with the for DNA isolation, the laboratory requested a redraw of the
proteolytic enzyme trypsin. specimen.
2. The mutation is an example of a structural rearrangement 6. After receiving the correct specimen for DNA isolation, the
between chromosomes 9 and 22, called the Philadelphia molecular scientist conducted the test; the result was the gel
chromosome. The Philadelphia chromosome represents a pattern seen in Figure 32-1. However, the supervisor deter-
balanced translocation between the long arms of chromo- mined that this gel electrophoresis result was not acceptable
somes 9 and 22. At the molecular level, the gene for ABL, and that the procedure had to be repeated. The reason for
an oncogene, joins a gene on chromosome 22 named BCR. the rejection is the lack of a no-DNA control. The no-DNA
The result of the fusion of these two genes is a new fusion control is essential when any polymerase chain reaction
protein. (PCR) test is performed in the clinical laboratory. This
3. Fluorescence in situ hybridization (FISH) is a molecular control will demonstrate whether cross-contamination
technique that uses DNA or RNA probes labeled directly occurred during the setup of the PCR procedure. The
with a fluorescent nucleotide or with a hapten (e.g., dini- no-DNA control region of the gel should lack a banding
trophenyl, digoxigenin, or biotin). Both the probe and pattern, as seen in Figure 32-2. If a banding pattern is
either metaphase or interphase cells are made single- present in the no-DNA control region or this control is
stranded (denatured) and then hybridized together. Cells missing, the test must be repeated before reporting patient
hybridized with a direct-label probe are viewed with a results.
fluorescence microscope. If the probe was labeled with a 7. Examination of the patients results in Figure 32-2 shows
hapten, antibodies to the hapten, carrying a fluorescent tag, that three bands (141, 104, 82bp) are present, which dem-
are applied to the cells. Once the antibodies bind to the onstrates that this patient is heterozygous for the factor V
RNA or DNA probe, the cells can be viewed using a fluores- Leiden mutation.
cence microscope. FISH complements standard chromo-
some analysis by confirming the G-band analysis and by Review Questions
improving resolution, which allows for analysis at the 1. d
molecular level. 2. b
3. d
Review Questions 4. b
1. c 5. b
2. d 6. c
3. a 7. a
4. d 8. a
5. a 9. b
6. c 10. b
7. d
8. c
CHAPTER 33
9. c
10. b Case Studies
Case 1
1. The lymphoid population is the most prominent. Forward
CHAPTER 32
scatter demonstrates small to medium-sized cells. These
Case Study cells are characterized by low side scatter indicative of sparse
1. DNA isolation for the detection of inherited mutations agranular cytoplasm.
requires whole blood collected in a lavender-topped tube 2. The majority of cells express CD19, CD10, and light chain.
containing EDTA to preserve white blood cells. There is also a small population of T cells positive for CD5
2. In the first gel, the correct controls are not present because and negative for CD19 antigen.
it lacks a no-DNA control. 3. Prominent light chain expression indicates a monoclonal
3. Bands in the patients sample appear at 141, 104, and 82bp. B-cell population that is characteristic of lymphoma.
APPENDIX Answers 811
Case 2 6. First-line therapy for CML is the tyrosine kinase inhibitor
1. The low density of CD45 antigen coupled with relatively imatinib mesylate (Gleevec). Allogeneic stem cell transplan-
low side scatter is characteristic of a blast population. Such tation should be considered for all CML patients, because
a prominent blast population can only be seen in acute it is the only potentially curative treatment for CML.
leukemias. However, few CML patients qualify for allogeneic stem cell
2. The expression of immature markers (CD34 and HLA-DR) transplantation, because most do not meet the criteria for
coupled with positivity for myeloid and megakaryoblastic low risk: age younger than 40 years of age, disease in the
antigens (CD33, CD41, and CD61) is seen in acute mega- chronic phase, transplantation within 1 year of diagnosis,
karyoblastic leukemias. and availability of an HLA-matched donor. For those
patients who qualify for allogeneic stem cell transplanta-
Review Questions tion, imatinib is used to induce remission prior to trans-
1. c plant, to treat minimum residual disease, and to provide
2. b rescue therapy if the transplant fails. Imatinib is continued
3. a as lifelong therapy until drug resistance is detected.
4. d 7. The majority of cases of imatinib resistance result from
5. b two primary causes: acquisition of additional BCR/ABL
6. a mutations and expression of point mutations in the adenos-
7. a ine triphosphate (ATP) binding site. Additional BCR/ABL
8. a mutations can occur through the usual translocation of the
9. c remaining unaffected chromosomes 9 and 22, which con-
10. a verts the hematopoietic stem cell from heterozygous to
11. b homozygous for the BCR/ABL mutation. A double dose of
BCR/ABL can also be acquired from gene duplication during
mitosis and accounts for 10% of secondary mutations. An
CHAPTER 34
additional BCR/ABL mutation will double the tyrosine
Case Study kinase activity, which makes the imatinib dosage inade-
1. An elevated white blood cell (WBC) count with a left shift quate. In these cases higher dosages of imatinib will restore
suggests a myeloproliferative neoplasm or a leukemoid remission in most patients. Over 50 mutations have been
reaction. However, in this patient the WBC count was identified in the ATP binding site, and these account for the
extremely elevated, the left shift was rather deep (presence remaining 50% to 90% of secondary mutations. Mutations
of promyelocytes and blasts), and basophilia was present, in the ATP binding site reduce the binding affinity of ima-
which suggests that a myeloproliferative neoplasm is likely tinib, producing some level of resistance.
present. Because two or more cytopenias were not evident
and teardrop cells were not reported, chronic myelogenous
leukemia (CML) is the most likely cause of these laboratory Review Questions
findings. 1. b
2. The leukocyte alkaline phosphatase (LAP) score is low in 2. c
CML due to inappropriate LAP synthesis in the secondary 3. d
granules, whereas LAP is elevated in bacterial infections due 4. c
to activation of enzyme synthesis. 5. c
3. The BCR/ABL1 fusion gene must be identified to confirm 6. c
the diagnosis of CML. BCR/ABL1 can be demonstrated from 7. b
a karyotype analysis showing the t(9;22) reciprocal translo- 8. d
cation know as the Philadelphia chromosome or by demon- 9. a
stration of the BCR/ABL1 fusion gene using fluorescence in 10. c
situ hybridization or molecular techniques. Patients who
have complete blood count and differential results that
resemble those in CML but who test negative for BCR/ABL1
CHAPTER 35
are considered to have atypical CML, and the disorder is
classified as myelodysplastic syndrome/myeloproliferative Case Study
neoplasm. 1. The differential diagnosis of patients with pancytopenia
4. Cytogenetic studies are likely to show the t(9:22) should include megaloblastic anemia (vitamin B12 or folate
mutation. deficiency), aplastic anemia, liver disease, alcoholism, and
5. The t(9;22) translocation produces the BCR/ABL1 chimeric myelodysplastic syndrome (MDS).
gene, which is observed in four primary molecular forms 2. The probable diagnosis is MDS.
that produce three versions of the BCR/ABL chimeric 3. This patients MDS should be classified as refractory anemia
protein: p190, p210, and p230. with ringed sideroblasts (RARS).
812 APPENDIX Answers

Review Questions Review Questions


1. d 1. d
2. a 2. c
3. b 3. d
4. b 4. d
5. c 5. b
6. c 6. b
7. a 7. b
8. d 8. a
9. c 9. c
10. c 10. a

CHAPTER 36 CHAPTER 38
Case Study Case Study
1. This is a case of acute lymphoblastic leukemia (ALL). The 1. Yes, typically the hemoglobin is between 16.5 and 21.5g/
chronic leukemias or myeloproliferative neoplasms usually dL and the hematocrit is 48% to 68%.
occur in adults and are rarely associated with thrombo 2. These values are normal for newborns. Erythrocytes of a
cytopenia at presentation. The blasts pictured are small newborn are markedly macrocytic. There may be up to 10
lymphoblasts. nucleated red blood cells on the first postnatal day, but they
2. This child has clinical features indicative of a good progno- disappear by day 5. The polychromasia reflects the reticulo-
sis: young age, female gender, and low white blood cell cytosis that persists for about 3 days.
count. A poor prognostic finding is the headache, which 3. These values are normal for newborns. The white blood cell
may represent central nervous system involvement. count of a newborn fluctuates a great deal, and leukocytosis
3. Hyperdiploidy carries a favorable prognosis in B-cell ALL in without evidence of infection is common. The differential
children, but a poor prognosis in adult ALL. may show an increase in neutrophils rather than the
lymphocyte predominance seen after 2 weeks. In this case
Review Questions the neutrophils and lymphocytes were present in equal
1. b amounts, but no immature neutrophils were seen.
2. b
3. a Review Questions
4. d 1. d
5. b 2. b
6. d 3. a
7. c 4. b
8. c 5. c
9. b 6. c
10. b 7. d
8. b
9. a
10. c
CHAPTER 37
Case Study
1. Diffuse large B-cell lymphoma (DLBCL).
CHAPTER 39
2. This lesion is expected to show exclusive (clonal) or
light chain expression. Flow cytometry is particularly sensi- Review Questions
tive in detecting surface and cytoplasmic immunoglobulin 1. a
light chains and is commonly used to confirm clonality of 2. c
lymphoproliferative disorders. In addition, other panB- Summary of questions 1 and 2
cell markers can be studied by flow cytometry (e.g., CD19,
CD22, and FMC7 antigens) to demonstrate the B-cell origin Instrument NRBCs Blasts
of this lymphoma. ADVIA +++ +
3. Most often DLBCL presents as a localized disease involving CELL-DYN NRBC No flag
a group of lymph nodes. Bone marrow involvement is rare
Coulter No result No flag
at presentation; however, it can occur later in the course of
the disease. Sysmex NRBC? No flag
APPENDIX Answers 813

3. c In advanced liver disease, poor liver circulation causes


4. Yes, a manual differential should be performed based on pressure in the portal circulation. This enlarges the spleen
the presence of nucleated red blood cell (NRBC) and blast (splenomegaly). The enlarged spleen sequesters and clears
flags. The low platelet count should also be verified during platelets more rapidly than normal, a condition called hyper-
the blood film review. splenism, which causes thrombocytopenia. In most cases,
5. c platelet function is reduced. This reduced platelet function
6. c can be demonstrated in the laboratory using platelet
7. b aggregometry and is the reason for the patients epistaxis.
8. a Vitamin K deficiency is also a possibility. To differentiate
9. b vitamin K deficiency from liver disease, a factor V and VII
10. b activity assay is performed. In vitamin K deficiency factor
11. c VII activity is reduced but factor V activity is normal. In liver
12. d disease, both are reduced.
13. a 2. In early liver disease the vitamin Kdependent factors II
14. b (prothrombin), VII, IX, and X are produced with diminished
15. c function. This can be corrected with a trial dose of oral or
intravenous vitamin K. In people with true vitamin K defi-
ciency secondary to an altered diet, the vitamin K therapy
CHAPTER 40
corrects bleeding and normalizes the PT and PTT, but in
Case Study liver disease vitamin K may not have a lasting effect. This is
1. Given the family history, this may be an inherited condi- because the liver cannot process the vitamin K normally.
tion, although pregnancy is an independent risk factor for In addition to vitamin K therapy, thawed frozen plasma
thrombosis. (FP) transfusion at 1 to 2 units/day is effective in supple-
2. Thrombosis is probably caused by the deficiency of a menting the livers production of all the coagulation factors.
coagulation inhibitor such as protein C, protein S, or anti- Cryoprecipitate may also be used to raise the fibrinogen
thrombin. It may be caused by a procoagulant gain-of- concentration, and platelet concentrate may be used if the
function mutation such as the factor V Leiden mutation or platelet count drops to below 50,000/mcL and there is con-
the prothrombin G20210A mutation. tinued evidence of mucocutaneous bleeding.
Administration of vitamin K, FP, cryoprecipitate, and
Review Questions platelets does not cure liver disease; these therapies only
1. b treat the bleeding symptoms. Additional treatment may
2. c include antibiotics, antivirals, and antiinflammatory drugs.
3. b
4. d Review Questions
5. b 1. a
6. d 2. d
7. b 3. b
8. c 4. c
9. a 5. b
10. a 6. a
7. d
8. d
CHAPTER 41
9. a
Case Study 10. a
1. The combination of thrombocytopenia and prolonged 11. c
prothrombin time (PT) and partial thromboplastin time
(PTT) indicate probable liver disease. In the absence of
CHAPTER 42
a full medical history, the patients hemarthroses and
description of himself as a bleeder leads to the pre- Case Study
sumption of hemophilia, possibly hemophilia A. It is pos- 1. The following tests for congenital and acquired risk factors
sible that he contracted hepatitis C from an untreated are included in a thrombophilia profile. Results for the
blood product. Treatment of factor concentrates for viral items with asterisks are valid only when the test is per-
disease began in 1984. Prior to 1984 most hemophilia formed 10 to 14 days after termination of antithrombotic
patients eventually developed hepatitis B or C from factor therapy or resolution of a thrombotic event.
concentrates. Hepatitis A is also a possibility. Liver disease Homocysteine
may be confirmed using bilirubin and liver enzyme Lupus anticoagulant profile*
assays. Prothrombin G20210A mutation
814 APPENDIX Answers

Activated protein C resistance* 3. b


Factor V Leiden mutation (confirmatory for activated 4. c
protein C resistance) 5. b
Anticardiolipin antibodies by immunoassay 6. b
Protein C functional assay and follow-up immunoassay* 7. b
Protein S functional assay and follow-up immunoassay* 8. c
Antithrombin functional assay and follow-up 9. d
immunoassay* 10. a
2. The most common acquired thrombotic risk factors are
antiphospholipid antibodies and lupus anticoagulant, and
CHAPTER 44
these are most often implicated in a thrombotic event.
3. Patients with thrombotic risk factors may be instructed to Case Study
avoid situations and practices that may trigger thrombosis, 1. Storage pool disease, aspirin-like defects, and use of anti-
such as immobilization, smoking, and use of oral contra- platelet agents such as aspirin are possibilities.
ceptives or hormone replacement therapy. They may be 2. Storage pool disease or aspirin-like defects seem most likely.
provided with prophylactic antithrombotic therapy at times 3. Based on the results of the quantitative test for adenosine
when circumstances increasing thrombotic risk cannot be triphosphate release, the likely cause is -granule storage
avoided, such as when undergoing orthopedic surgery. pool disease.
These results were confirmed by the findings of electron
*Valid only when performed 10 to 14 days after termination of anti- microscopy of the patients platelets, which revealed the
thrombotic therapy or resolution of a thrombotic event. absence of detectable -granules. Because the patients bleed-
ing problems are due to an inherited abnormality that typi-
Review Questions cally results in only mild bleeding problems, the patient was
1. a counseled to avoid antiplatelet agents, particularly aspirin,
2. a since they are known to exacerbate the bleeding problems
3. d encountered by patients with -granule deficiency.
4. c
5. b Review Questions
6. a or b 1. d
7. a 2. a
8. a 3. a
9. d 4. b
10. d 5. c
11. b 6. c
12. d 7. d
13. d 8. b
14. c 9. d
10. a

CHAPTER 43
CHAPTER 45
Case Study
1. Yes, the heparin is significant. Case Study
2. Heparin-induced platelet aggregation assay, serotonin 1. The laboratory director questioned the phlebotomist about
release assay, or enzyme-linked immunosorbent assay the problem. The phlebotomist admitted that he had erro-
(ELISA) should be ordered. neously collected blood in a red- and gray-stoppered tiger-
An ELISA was performed to look for the presence of top tube and, responding to the patients remark, had
heparin-induced antibodies. Results gave an optical density immediately poured the blood into a blue-stoppered tube
of 0.50, with a reference range of less than 0.400 optical for analysis. He thought the specimen would be okay
density. The patient was found to have clinically significant because it had not clotted yet.
levels of heparin-induced antibodies. 2. The red and gray marbleized stopper designates a serum sepa-
The patient underwent an above-knee amputation of her rator tube. The phlebotomist poured the blood into the blue-
right leg. The grafting surgeries were successful, and the stoppered tube before it had begun to clot; however, the
patient recovered nicely. activator from the tiger-top tube shortened the clotting time
on the prothrombin time (PT) test, thus causing an errone-
Review Questions ously short PT and low international normalized ratio (INR).
1. b 3. Unexpectedly short PTs during oral anticoagulant therapy
2. d are generally indicators of patient non-compliance to the
APPENDIX Answers 815

drug regimen. The second most common circumstance that give vitamin K orally or intravenously to stop bleeding if
affects the PT is dietary changes, most often an increased necessary.
intake of vitamin Krich foods such as green leafy vegetables, 3. The chromogenic factor X assay.
liver, or avocado. In this instance the patient had been fully
compliant, carefully following the prescribed dosage and Review Questions
timing, and her diet had not changed. These facts led the 1. c
laboratory director to consider a specimen collection error. 2. c
Specimens collected in 3.2% sodium citrate may be 3. c
stored for up to 24 hours at room temperature without a 4. a
change in the PT. However, specimens stored at higher than 5. d
24 C deteriorate rapidly, which causes prolongation of the 6. b
PT and increase in the INR. Prolonged storage at 2 to 7. d
4 C may activate factor VII, which slightly shortens the PT 8. c
and slightly decreases the INR. 9. b
Many serum separator tubes contain particulate materi- 10. b
als that hasten in vitro clotting. Core laboratory managers 11. a
select these tubes to improve test result turnaround time 12. a
when the required sample is serum. When blood is col- 13. a
lected into a series of tubes that includes a blue-stoppered 14. d
tube for hemostasis testing, the blue-stoppered tube should 15. d
be filled first or should be filled after a tube without addi-
tives. It should not be filled immediately after filling a
CHAPTER 47
serum separator tube with clot activators, because the acti-
vators may carry over to the hemostasis specimen and affect Case Study
test results. 1. No. The description of the sample and the instrument flags
In this case, an observant patient provided clues that led indicating lipemia should alert the operator to a potentially
to identification of the preanalytical error. The phleboto- invalid test result, because lipemia is known to cause erro
mist was carefully counseled about the effects of tube addi- neous results on some photo-optical coagulation analyzers.
tives on hemostasis tests. 2. Two options are available to negate the effect of the lipemia
and obtain valid test results:
Review Questions a. Remove the lipids from the plasma by high-speed
1. c centrifugation or ultracentrifugation.
2. b b. Perform testing using an end-point detection method
3. a that is not susceptible to lipemia in the sample, such
4. b as mechanical clot detection.
5. a 3. Because the patient history includes previous surgical
6. a procedures without bleeding symptoms and there is no
7. b other indication of abnormal bleeding tendencies for this
8. b patient, it is probably safe to consider that the prolonged
9. b prothrombin time and activated partial thromboplastin
10. d time results are due to the lipemic nature of the sample.
11. b The patient would most likely not be at risk for bleeding
12. b during the surgery, and it would be anticipated that repeat
13. a or d testing using one of the options listed previously would
14. d yield test results within the reference range.
15. c
Review Questions
1. b
CHAPTER 46
2. d
Case Study 3. a
1. The increase in anticoagulation could be caused by a change 4. c
in diet, dietary supplements, or drugs. Any new drug that 5. b
interferes with the cytochrome oxidase P-450 enzyme 2C9 6. c
pathway could reduce warfarin breakdown and excretion, 7. c
and increase its effectiveness. 8. a
2. Determine what has caused the change in warfarin efficacy 9. d
and eliminate it if possible, adjust the warfarin dosage, or 10. a
Glossary
abetalipoproteinemia Autosomal recessive disorder of acrocyanosis (Raynaud phenomenon) Persistent sym-
lipoprotein metabolism in which lipoproteins containing metrical cyanosis (blotchy blue or red discoloration) of the
apolipoprotein B (chylomicrons, very-low-density lipopro- skin of the digits, palm, wrists, and ankles upon prolonged
teins, and low-density lipoproteins) are not synthesized. exposure to cold.
Characterized by the presence of acanthocytes on a peri activated coagulation time (ACT) Whole-blood clotting
pheral blood film and low levels of cholesterol in the time test often used in cardiac surgical suites. A particulate
plasma. activator is added to blood, the mixture is rocked, and the
absolute neutrophil count (ANC) Total neutrophils per interval to clotting is recorded. Used to monitor high-dose
liter of blood. The absolute neutrophil count is calculated unfractionated heparin therapy.
by multiplying the patients total white blood cell count by activated partial thromboplastin time (APTT, partial
the percentage of segmented neutrophils and bands or may thromboplastin time, PTT) Clot-based screening test
be counted directly using an automated hematology for intrinsic coagulation, which is prolonged in deficiencies
analyzer. of prekallikrein, high-molecular-weight kininogen, and
absolute reticulocyte count (ARC) Reticulocytes per factors XII, XI, IX, VIII, X, V, II, and I. Calcium chloride,
liter. The absolute reticulocyte count is calculated by multi- phospholipid, and activator are added to patient plasma.
plying the patients visual reticulocyte count (reticulocyte The interval from the addition of reagent to clot formation
percentage, or VRET%) by the red blood cell count or may is recorded. The test is used to monitor unfractionated
be measured directly using an automated hematology heparin therapy and to screen for intrinsic pathway deficien-
analyzer. cies, specific factor inhibitors, and lupus anticoagulant.
acanthocyte Red blood cell with spiny projections of varying activated protein C (APC) Coagulation pathway regulatory
lengths distributed irregularly over its surface, associated protein activated by the thrombin-thrombomodulin com
with lipid imbalance. Contrast with the echinocyte, which plex that, when bound and stabilized by protein S, hydro-
has regular projections of uniform length. lyzes and inactivates factor Va and factor VIIIa.
acanthocytosis Presence of acanthocytes in the blood. Asso- activated protein C resistance (APCR) Inherited condi-
ciated with abetalipoproteinemia or abnormalities of lipid tion in which activated coagulation factor V (Va) resists
metabolism, such as abnormalities occurring in liver disease. activated protein C digestion, which results in an increased
accuracy Extent to which an assay result matches its true risk of venous thrombosis. In 90% of cases, activated protein
value. Accuracy is computed by comparing assay results with C resistance is caused by the factor V Leiden mutation.
the results from an established reference assay or a primary activated prothrombin complex concentrate Therapeu-
standard. tic plasma preparation that bypasses factor VIII activation,
achlorhydria Pathologic absence of free hydrochloric acid used to treat bleeding episodes in hemophilic patients
from gastric secretions following stimulation. who have developed factor VIII inhibitor. Contains acti-
acid elution slide test (Kleihauer-Betke stain) Test for vated factors II (prothrombin), VII, IX, and X. Also known
detecting fetal red blood cells (RBCs) in the maternal circu- as prothrombin complex concentrate, factor eight inhibitor
lation. Blood films are immersed in an acid buffer, which bypassing activity (FEIBA; Baxter Healthcare Corporation,
causes adult hemoglobin (Hb A) to be eluted from RBCs. Deerfield, Ill).
The film is stained, and RBCs that have fetal hemoglobin acute Describes diseases whose symptoms begin abruptly
(Hb F) take up the stain. with marked intensity and then subside after a relatively
acquired immunodeficiency syndrome (AIDS) Late- short period.
stage immune system suppression characterized by deple- acute leukemia Malignant, unregulated proliferation of
tion of CD4+ T lymphocytes and depression of cellular hematopoietic progenitors of the myeloid or lymphoid cell
immunity causing susceptibility to opportunistic infections lines. Characterized by abrupt onset of symptoms and, if left
and neoplasms. Caused by infection with human immuno- untreated, death within months of the time of diagnosis.
deficiency virus (HIV), a retrovirus. acute myocardial infarction (AMI) Occlusion of a coro-
acrocentric Describes the appearance of a metaphase chro- nary artery by a clot, causing ischemia and necrosis (tissue
mosome with the centromere near one end, which causes death) of surrounding heart muscle. Commonly called a
the q arm to be much longer than the p arm. heart attack.

816
APPENDIX Glossary 817
acute phase reactant Serum protein produced by the liver of circulating RBCs. This is the basis for hemolytic disease
whose level rises during inflammation. Examples include of the newborn.
C-reactive protein, ferritin, and fibrinogen. -granules Platelet granules that store and release a variety
adenopathy Inflammatory enlargement of a lymph gland. of hemostasis proteins. There are 50 to 80 -granules per
adenosine diphosphate (ADP) Purine nucleoside that platelet. In transmission electron microscopy, -granules
activates platelets by binding platelet receptor P2Y1 and appear light gray, in contrast to -granules (dense bodies),
P2Y12. Produced by hydrolysis of adenosine triphosphate. which appear black.
adenosine triphosphate (ATP) Purine nucleoside that -thalassemia Moderate to severe inherited anemia caused
stores energy in the form of high-energy phosphate bonds, by a decreased rate of synthesis of the -globin chains of
releasing energy upon hydrolysis to drive metabolic hemoglobin.
reactions. amyloidosis Disease in which a waxy, starchlike glycoprotein
adhesion Property of binding or remaining in proximity; for (amyloid) accumulates in tissues and organs, impairing
example, attachment of platelets to surfaces such as suben- their function.
dothelial collagen. analogue drugs Drugs that are chemically similar to one
adipocyte Fat cell; adipocytes make up adipose tissue and the another but, because of minor structural differences, may
yellow portion of the bone marrow. have different physiologic actions.
afibrinogenemia Complete absence of plasma fibrinogen. anaplastic Characterized by loss of differentiation, and
agammaglobulinemia Immunodeficiency characterized by growth without structure or form. Anaplasia is a character-
an absence or extremely reduced level of plasma gamma istic of cancer.
globulin and reduced levels of immunoglobulins. Associ- anatomic bleeding disorder Chronic episodic bleeding
ated with increase risk of infection. into soft tissue, joints, and body cavities. Indicates a second-
agglutination Cross-linking of antigen-bearing cells or par- ary coagulopathy such as hemophilia or an acquired coagu-
ticles by a specific antibody to form visible clumps. lation factor deficiency.
aggregation Cluster or clump of similar cell types or parti- anemia Diminished delivery of oxygen to tissues, as evi-
cles; for example, attachment of platelets to other platelets, denced by pallor, malaise, and dyspnea. May be caused by
red blood cell clumping. blood loss, decreased red blood cell (RBC) production,
agnogenic Of idiopathic or unknown origin. increased RBC destruction (shortened life span), low iron
agranulocytosis Any condition involving decreased numbers concentration, or abnormal hemoglobin.
of granulocytes (segmented neutrophils or band neutrophils). aneuploid Having a chromosome number that is not an
albinism Hereditary condition characterized by partial or exact multiple of the normal diploid number (46 in
total lack of melanin pigment in the body; skin, hair, and/ humans) so that there are fewer or more chromosomes
or eyes may be affected. Individuals with total albinism than normal.
have pale skin that does not tan, white hair, and pink eyes. anisocytosis Abnormal red blood cell (RBC) morphology
Albinism is often associated with platelet storage pool characterized by considerable variation in RBC volume or
deficiency. RBC diameter on a blood film.
Alder-Reilly anomaly Autosomal dominant polysaccharide annular diaphragm Device used in phase microscopy that,
metabolism disorder in which white blood cells (WBCs) of together with a phase-shifting element, produces contrast
the myelocytic series, and sometimes all WBCs, contain between an unstained cell and its dark background.
coarse azurophilic mucopolysaccharide granules. anoxia Inadequate tissue oxygenation caused by poor lung
allele One of two or more alternative forms of a gene that perfusion or a diminished blood supply.
occupy corresponding loci on homologous chromosomes. antagonist Drug that nullifies the action of another drug or
Each allele encodes a certain inherited characteristic. An that reduces a normal cellular response. Aspirin is a platelet
individual normally has two alleles for each gene, one con- antagonist because it reduces platelet activation.
tributed by the mother and one by the father. If both alleles antecubital fossa Concavity opposite the elbow.
are the same, the individual is homozygous, but if the alleles antibody (Ab) Specialized protein (immunoglobulin) that
are different, the individual is heterozygous. In heterozy- is produced by B lymphocytes when the immune system is
gous individuals, one of the alleles may be dominant and exposed to foreign antigens from bacteria, viruses, or other
the other recessive. biologic materials. An antibody molecule has a specific
alloantibody (isoantibody) Antibody that is produced in amino acid sequence in its variable region that matches it
response to the presence of foreign antigens; for instance, to the antigen which originally stimulated its production.
an antibody to a therapeutic coagulation factor that may anticardiolipin antibody (ACL, ACA) Member of the
render factor therapy ineffective. antiphospholipid antibody family that includes anti2-
alloimmune Producing antibodies to antigens derived from glycoprotein 1 and lupus anticoagulant. An ACL antibody
a genetically dissimilar individual of the same species. is an autoantibody detected in a solid-phase immunoassay
alloimmune hemolytic anemia Anemia caused by anti- system using cardiolipin as the target antigen. The chronic
bodies stimulated by exposure to foreign red blood cell presence of ACL antibodies is associated with venous and
(RBC) antigens. Antibodies coat and shorten the life span arterial thrombotic disease.
818 APPENDIX Glossary

anticoagulant Therapeutic agent that delays blood coagula- development, a difference in rate between cytoplasmic and
tion, such as heparin or warfarin, used to prevent throm- nuclear maturation.
botic events in patients who are at risk. Additive to blood atrial fibrillation (AFIB) Uncontrolled and ineffective
specimen collection tubes that prevents in vitro blood atrial heartbeat that affects at least 2 million U.S. citizens.
clotting such as sodium citrate. Atrial fibrillation raises the risk for stroke. The risk is con-
antigen Molecule, usually a protein, that the immune system trolled using long-term warfarin therapy.
recognizes as foreign and that subsequently evokes an atypical lymphocytes (transformed, reactive, or variant
immune response. lymphocytes) Lymphocytes whose altered morphology
antihemophilic factor (AHF) Therapeutic concentration includes stormy blue cytoplasm and lobular or irregular
of coagulation factor VIII produced through chemical frac- nuclei. Variant lymphocytes indicate stimulation by a virus,
tionation, immunoaffinity column, or recombinant synthe- particularly Epstein-Barr virus, which causes infectious
sis. Antihemophilic factor is prescribed in the treatment of mononucleosis.
hemophilia A, a hereditary deficiency of factor VIII. Auer rod Abnormal needle-shaped or round pink to purple
antineoplastic Chemotherapeutic agent that controls or kills inclusion in the cytoplasm of myeloblasts and promyelo-
cancer cells. cytes; composed of condensed primary granules. Indicates
antiphospholipid antibody (APL, APA) Member of the acute myeloid leukemia.
antibody family that includes anticardiolipin, anti2- autoantibody Antibody produced by an individual that rec-
glycoprotein I, and lupus anticoagulant. APL antibodies ognizes and binds an antigen on the individuals own
bind phospholipid-binding proteins, such as 2- tissues.
glycoprotein. The presence of an APL antibody is associated autoimmune Describes an immune response in which an
with venous and arterial thrombotic disease. antibody forms to ones own tissues; for instance, antinu-
antiphospholipid syndrome (APS) Group of thrombotic clear antibody in lupus erythematosus.
disorders related to the chronic presence of an antiphospho- autoimmune hemolytic anemia Anemia characterized by
lipid antibody, such as anticardiolipin, anti2-glycoprotein premature red blood cell (RBC) destruction. Autoantibodies
I, or lupus anticoagulant. Manifestations include migraine, to RBC surface antigens bind the membrane, causing rapid
transient ischemic attacks, strokes, acute myocardial infarc- splenic clearance and hemolysis.
tion, peripheral artery disease, venous thromboembolic autologous Related to self or belonging to the same organ-
disease, and spontaneous abortion. ism; for example, used to describe blood that is donated by
antithrombin (AT, antithrombin III, AT III) Plasma patients before surgery for the purpose of transfusion to
serine protease inhibitor produced in the liver and activated themselves during or after surgery.
by therapeutic heparin or vascular heparan sulfate. When autosomal dominant inheritance Pattern of inheritance
activated, antithrombin controls the coagulation pathway, in which the transmission of a dominant allele on an auto-
because it neutralizes all the serine proteases except factor some causes a trait to be expressed in heterozygotes.
VIIa, most importantly factors IIa (thrombin) and Xa. autosomal inheritance Inheritance of traits located on
aperture Optical device in a microscope substage light path nonsex-related chromosomes.
that controls the diameter of the light column that reaches autosomal recessive inheritance Pattern of inheritance
the specimen. The aperture may be adjusted to improve resulting from the transmission of a recessive allele that is
specimen clarity. not expressed in heterozygotes.
aplasia Failure of the normal process of cell generation and autosome Any of the 22 pairs of chromosomes in humans
development. Bone marrow aplasia is the loss of all bone other than the sex chromosomes X and Y.
marrow cellular elements. autosplenectomy Disappearance of the spleen through pro-
aplastic anemia Deficiency of all of the formed elements of gressive fibrosis and shrinkage secondary to a hemolytic
blood, representing a failure of the blood cellgenerating anemia such as sickle cell anemia.
capacity of bone marrow. azurophilic Having cellular structures that stain blue with
apoferritin Protein component of the ferritin molecule con- Giemsa stain and red-purple with Wright stain.
sisting of 24 identical spherical subunits that surround iron azurophilic granules Primary cytoplasmic granules in
in the ferric valence form. myelocytic cells that, when stained with Wright stain, appear
apoptosis Natural cell death characterized by nuclear con- reddish purple. Azurophilic granules of different composi-
densation and loss of cytoplasmic integrity. Apoptosis is a tion may also appear in a minority of lymphocytes.
mechanism that prevents proliferation of dysplastic or B cell Any of a family of lymphocytes that produce antibod-
mutated cells. ies. The end product of B-cell maturation is the plasma cell.
aspirin (acetylsalicylic acid, ASA) Acetylsalicylic acid in Babesia Protozoal parasite transmitted by ticks that infects
tablet form; irreversibly acetylates platelet cyclooxygenase human red blood cells and causes babesiosis, a malaria-like
and reduces platelet activation. Aspirin is used for its anti- illness. The parasite is an intracellular ringlike structure 2 to
thrombotic properties. 3m in diameter.
asynchrony Disturbance of coordination that causes pro- band neutrophil (band) Immediate precursor of the
cesses to occur at abnormal times. In hematopoietic cell mature segmented neutrophil. Band neutrophils have a
APPENDIX Glossary 819

nonsegmented, usually curved, nucleus and are present in one of the main regulators of oxygen uptake and delivery
the bone marrow and peripheral blood. by hemoglobin. 2,3-BPG decreases hemoglobins affinity
bartonellosis Acute febrile infection caused by the bacterium for oxygen, which enables it to more readily release oxygen
Bartonella bacilliformis, which is transmitted by the bite of a to the target tissues.
sandfly. The first stage of the disease is associated with severe blast Earliest, least differentiated stage of hematopoietic mat-
hemolytic anemia. uration that can be identified by its morphology in a Wright-
base pair Pair of nucleotides in the complementary strands stained bone marrow smear; for example, myeloblast,
of a DNA molecule that interact through hydrogen bonding pronormoblast (rubriblast), lymphoblast.
across the axis of the DNA helix. One of the nucleotides in bleeding time (BT) Time interval required for blood to stop
each pair is a purine (either adenine or guanine), and the flowing from a puncture wound 2mm long and 1mm deep
other is a pyrimidine (either thymine or cytosine). Adenine on the volar surface of the forearm. Largely of historical
always pairs with thymine, and guanine always pairs with interest, the bleeding time test was performed to evaluate
cytosine. vascular and platelet function.
basophil (baso) Granulocytic white blood cell characterized Bohr effect Effect of carbon dioxide and hydrogen ions
by cytoplasmic granules that stain bluish black when on the affinity of hemoglobin for oxygen. Increasing
exposed to a basic dye. Cytoplasmic granules of basophils carbon dioxide and hydrogen ion concentrations (lower
are of variable size and may obscure the nucleus. pH) decrease oxygen saturation; decreasing concentrations
basophilia Abnormal increase of basophils in the blood. increase oxygen saturation.
basophilic normoblast (prorubricyte) Second identifi- bone marrow Gelatinous red and yellow tissue filling the
able stage in bone marrow erythrocytic maturation; it medullary cavities of bones. Red marrow is found in most
is derived from the pronormoblast (rubriblast). Typically bones of infants and children and in the ends of long bones
10 to 12m in diameter, the basophilic normoblast (pro- and the cavities of flat bones in adults. Diagnostic marrow
rubricyte) has cytoplasm that stains dark blue with Wright specimens are collected from the anterior or posterior iliac
stain. crest or sternum in adults. Fatty yellow marrow is found in
basophilic stippling Barely visible dark blue to purple gran- the medullary cavity of most adult long bones.
ules evenly distributed within a red blood cell stained with bone marrow aspirate specimen A 1- to 2-mL aliquot of
Wright stain. Composed of precipitated ribosomal protein gelatinous red marrow obtained by passing a needle into
and RNA. the marrow cavity and applying negative pressure. The aspi-
Bence Jones protein Protein found almost exclusively in rate specimen is spread as a smear on a microscope slide,
the urine of patients with multiple myeloma, consisting stained, and examined for hematologic or systemic disease.
of the light chain of the abnormal immunoglobulins The aspirate specimen provides for analysis of individual
produced. cell morphology.
benign Noncancerous or nonmalignant. bone marrow biopsy specimen A 1- to 2-mL cylinder
Bernard-Soulier syndrome (BSS) Mild to moderate of gelatinous red marrow obtained by passing a biopsy
mucocutaneous bleeding disorder caused by one of a series cannula into the marrow cavity, rotating, and withdrawing.
of mutations to platelet glycoprotein Ib (GP Ib) or GP IX, The cylinder is fixed in formalin, sectioned, stained,
part of the GP Ib/IX/V von Willebrand factor receptor and examined for hematologic or systemic disease. The
complex. The disorder is a defect of platelet adhesion. biopsy specimen provides for analysis of bone marrow
2-glycoprotein I (2-GPI) Plasma globulin that is a target architecture.
of the antiphospholipid antibody anti2-glycoprotein I. buffy coat Gray-white layer of white blood cells (WBCs) and
-thalassemia Inherited anemia caused by diminished syn- platelets that accumulates at the red blood cellplasma
thesis of the -globin chains of hemoglobin. interface when a tube of blood is allowed to stand or is
-thromboglobulin (-TG) Heparin-neutralizing protein centrifuged. The buffy coat may be used to harvest WBCs
that is stored and secreted by platelet -granules. for microscopic analysis when the WBC count is low, and
bilirubin Gold-red-brown pigment, the main component of an enlarged buffy coat may indicate leukemia.
bile and a major metabolic product of the heme portion of Burkitt lymphoma Lymphatic solid tissue tumor composed
hemoglobin, released from senescent red blood cells. Ele- of mature B lymphocytes with a characteristic morphology,
vated bilirubin imparts a gold color to plasma and urine called Burkitt cells. Burkitt cells appear in lymph node biop-
and may indicate hemolytic anemia, liver disease, or bile sies, bone marrow, and occasionally in peripheral blood
duct occlusion. and have dark blue cytoplasm with multiple vacuoles creat-
bilirubinemia (icterus, hyperbilirubinemia) Excess bili- ing a starry sky pattern.
rubin in plasma. It imparts a gold color to plasma and may burst-forming unit (BFU) Early hematopoietic progenitor
indicate hemolytic anemia, liver disease, or bile duct cell stages of the erythroid and megakaryocytic cell lines
occlusion. characterized by their tissue culture growth pattern in which
2,3-bisphosphoglycerate (2,3-BPG, 2,3-diphosphogly large colonies are produced. Contrast with the more dif-
cerate, 2,3-DPG) Product of red blood cell glycolysis that ferentiated colony-forming units, which produce smaller
is generated in the Rapoport-Luebering shunt. 2,3-BPG is colonies.
820 APPENDIX Glossary

C banding In cytogenetic analysis, a specialized Giemsa stain the sputum of asthma patients and the feces of dysentery
technique employing first an acid, then a basic buffer, that patients. Formed from the granules of disintegrating
highlights the centromeres of chromosomes. The stained eosinophils.
centromere is the C band, which helps to identify the Chdiak-Higashi anomaly Autosomal recessive disorder
chromosome. characterized by partial albinism, photophobia, susceptibil-
Cabot rings Threadlike structures that appear as purple-blue ity to infection, and the presence of giant blue granules in
loops or rings in Wright-stained red blood cell cytoplasm. Wright-stained white blood cells and platelets.
They are remnants of mitotic spindle fibers that indicate chelation Chemical formation of a ring-shaped molecular
hematologic disease such as megaloblastic or refractory complex in which a metal ion is covalently bound. Chelat-
anemia. ing agents such as ethylenediaminetetraacetic acid (EDTA)
calibrator (secondary standard) Preserved material in or sodium citrate are used as blood specimen anticoagu-
which the analyte concentration has been assigned by refer- lants. Chelators are also used to treat lead poisoning or iron
ence to a primary standard or by controlled reference assays overload.
in expert laboratories. Calibrators are used for assays in chemotaxis Cellular movement toward or away from a chem-
which there are no primary standards, such as blood cell ical stimulus. Characteristic of neutrophils and monocytes,
counts or coagulation assays. whose phagocytic activity is influenced by chemical factors
carboxyhemoglobin Hemoglobin that has bound carbon released by invading microorganisms.
monoxide, which prevents normal oxygen exchange. Car- chemotherapy Treatment of neoplastic disease (cancer) by
boxyhemoglobin imparts a cherry-red color to venous chemical agents.
blood, and its reduced oxygen capacity is the basis for chromogen Chemical that absorbs light and produces
carbon monoxide poisoning. color. Chromogens are used in laboratory analysis by
carcinoma Malignant neoplasm of epithelial cell origin that spectrophotometry.
invades surrounding tissue and may metastasize to distant chromophore The portion of a molecule that absorbs inci-
regions of the body. dent light and emits colored light, usually as a result of the
cell membrane Cell surface composed of two layers of phos- presence of 10 or more double bands or aromatic rings.
pholipids intermixed with cholesterol and a variety of spe- Chromophores are the colored portions of chromogens and
cialized glycoproteins that support cell structure, signaling, are synthesized in molecules to provide measurable color
and ion transport. in laboratory assays.
cellular immunity (cell-mediated immunity, CMI) chromosome Threadlike nuclear structure composed of con-
Immune response initiated and mediated by T-lymphocyte densed DNA that transmits genetic information. In humans
secretions called cytokines, natural killer cells, and macro- there are 46 chromosomes, including 22 homologous pairs
phages; the mechanism of acquired immunity characterized of autosomes and 1 pair of sex chromosomes, X and Y.
by the dominant role of the T lymphocytes. Cellular immu- chronic Persisting over a long period of time, often for the
nity is the basis for graft rejection, delayed hypersensitivity, remainder of a persons life.
and responses to viral infections and tumors. chronic leukemia Malignant, unregulated proliferation of
centriole Cylindrical nuclear organelle composed of micro- myelocytic or lymphocytic cells that appear at several stages
tubules. During mitosis the centriole replicates and the two of differentiation in peripheral blood. Characterized by
centrioles move to opposite ends of the cytoplasm, where slow onset and progression of symptoms.
they bind to spindle fibers that attach to the centromeres Clinical and Laboratory Standards Institute (CLSI)
of chromosomes and effect their movement during Global nonprofit agency that uses consensus to develop and
metaphase. publish healthcare guidelines and standards.
centromere Constricted portion of a chromosome that Clinical Laboratory Improvement Amendments (CLIA)
attaches to a spindle fiber to effect movement during meta- of 1988 Law establishing the Clinical Laboratory Improve-
phase. Centromeres are categorized by their location as ment Amendments Committee (CLIAC), which sets and
acrocentric (near one end), metacentric (near the center), enforces standards for quality testing in the clinical
or submetacentric (off center). laboratory.
cerebrospinal fluid (CSF) Fluid that flows through and clone Group of genetically identical cells derived from a
protects the four ventricles of the brain, the subarachnoid single common cell through mitosis.
spaces, and the spinal canal. CSF is derived from plasma Clostridium perfringens Anaerobic gram-positive bacteria
and is the site of bacterial and viral infections called that cause gangrene in humans. Symptoms of gangrene
meningitis or encephalitis. CSF is sampled by lumbar include intravascular hemolysis and thrombosis.
puncture. cluster of differentiation (CD) Cell surface membrane
cerebrovascular accident (CVA) Stroke; occlusion of an receptors or markers used to characterize cells by their func-
artery of the brain or brain hemorrhage resulting in necrosis tions. CD profiles are used in flow cytometry to identify cell
of brain tissue and loss of function. types. CDs are used in hematology to identify cell clones
Charcot-Leyden crystals Crystalline structures that are associated with lymphatic and myelogenous leukemias and
shaped like narrow double pyramids and are found in lymphomas.
APPENDIX Glossary 821
coagulation Series of enzymatic reactions beginning with and involves factors X, V, II (prothrombin), and fibrinogen,
activation of factor VII by tissue factor (extrinsic/in vivo) or in order of reaction.
factor XII by a negatively charged surface (intrinsic/in vitro) complement (C) System of at least 20 serum enzymes. The
and proceeding through the common pathway to the for- complement system responds to antigen-antibody reaction
mation of an insoluble fibrin clot. to stimulate white blood cell chemotaxis, generate inflam-
coagulation cascade Series of enzymatic reactions begin- mation, and cause red blood cell lysis.
ning with activation of factor VII by tissue factor (extrinsic/ condenser Substage microscope device that focuses light on
in vivo) or factor XII by a negatively charged surface (intrin- the slide-mounted specimen to promote visual clarity.
sic/in vitro) and proceeding through the common pathway confidence interval (CI) Range of values expected to
to the formation of an insoluble fibrin clot. contain the measured value (parameter) with a predeter-
coagulation factors Plasma proteins, also called procoagu- mined degree of statistical confidence. For instance, the 95%
lants, that circulate as inactive forms. When activated in the confidence interval is expected to include 95% of all values
process of coagulation, procoagulants participate in the of a parameter measured in a normal population, which
coagulation cascade to form a fibrin clot. corresponds closely to 2 standard deviations.
codocyte (target cell) Poorly hemoglobinized red blood congenital Describes a condition that exists at, and presum-
cell (RBC) that appears in hemoglobinopathies, thalasse- ably before, birth. Often refers to a hereditary condition.
mia, and liver disease. In a Wright-stained peripheral blood Coombs test (direct antiglobulin test, DAT) Screening
film, hemoglobin concentrates in the center of the RBC and procedure in which antihuman globulin is used to detect
around the periphery, causing the cell to resemble a antibodies and complement bound to red blood cells
bulls-eye. in vivo.
coefficient of variation (CV, percent CV, CV%) Statisti- coronary artery bypass grafting (CABG) Cardiac surgery
cal measure of the deviation of a variable from its mean usually requiring cardiopulmonary bypass and extracorpo-
divided by the mean, usually expressed as a percentage. real circulation in which occluded sections of coronary
colchicine Alkaloid that blocks microtubule formation arteries are replaced with grafts taken from nearby arteries,
and prevents cell division. Used in cytogenetic studies to for example, the internal mammary artery.
arrest mitosis in metaphase so that chromosomes may corrected reticulocyte count Calculation performed to
be karyotyped. Colchicine is also a component of a drug correct the visual reticulocyte count for specimens with a
used to treat gout. Colcemid is the trademark for a col hematocrit below 45% to the equivalent reticulocyte count
chicine derivative used in preparing chromosomes for at a hematocrit of 45%. In anemia, the visual reticulocyte
karyotyping. percentage is misleadingly elevated because whole blood
cold agglutinin IgM autoantibody specific for red blood cell contains fewer red blood cells relative to reticulocytes.
surface membrane antigens of the Ii system that reacts Coumadin (warfarin, oral anticoagulant therapy,
at temperatures below 30 C to cause cold agglutinin OAT) Vitamin K antagonist used as an anticoagulant
disease. to prevent thrombosis in people with atrial fibrillation,
cold agglutinin disease Acquired autoimmune hemolytic venous thromboembolism, or cardiac insufficiency. Also
anemia resulting from red blood cell (RBC) agglutination used prophylactically after orthopedic surgery. Warfarin
by IgM autoantibodies that coat RBCs at temperatures suppresses vitamin K and reduces the activity of the vitamin
below 30 C. Patients with cold agglutinin disease experi- Kdependent coagulation factors II (prothrombin), VII, IX,
ence Raynaud phenomenon, characterized by pallor, cyanosis, and X.
and pain in the fingers, palms, wrists, and ankles after expo- cryoglobulin Any of numerous serum globulins, typically
sure to cold. immunoglobulins, that precipitate at around 4 C and
colony-forming unit (CFU) Hematopoietic progenitor become resuspended at 37 C.
cells that are derived from the pluripotential hematopoietic cryoprecipitate (CRYO) Therapeutic agent rich in fibrino-
stem cell and give rise to the different cell lineages of the gen, factor VIII, and factor XIII, used to treat bleeding dis-
bone marrow. Named because of their ability to form colo- orders in fibrinogen deficiency, factor XIII deficiency, and
nies in tissue culture. hemorrhagic trauma. Cryoprecipitate is collected from
colony-forming unitgranulocyte, erythrocyte, mono- human plasma that has been frozen and slowly thawed.
cyte, and megakaryocyte (CFU-GEMM) Hemato cyanosis Bluish discoloration of the skin, sclera and mucous
poietic progenitor cell capable of differentiating into the membranes caused by poor tissue oxygenation. Usually a
granulocytic (myelocytic), erythrocytic (normoblastic), sign of anemia.
monocytic, or megakaryocytic cell lines. cytochemical analysis Use of specialized stains to detect
colony-stimulating factor (CSF) Cytokine that promotes cellular enzymes and other chemicals in peripheral blood
the division and differentiation of hematopoietic cells. films and bone marrow aspirate smears. Used to differenti-
common coagulation pathway The steps in the coagula- ate hematologic diseases, especially leukemias.
tion cascade from the activation of factor X through the cytogenetics Branch of genetics devoted to the laboratory
conversion of fibrinogen to fibrin. The common pathway study of visible chromosome abnormalities, such as dele-
begins at the junction of the intrinsic and extrinsic pathways tions, translocations, and aneuploidy.
822 APPENDIX Glossary

cytokine Cellular product that influences the function or adenine, cytosine, guanine, and thymine. During mitosis,
activity of other cells. Cytokines include colony-stimulating DNA condenses to form chromosomes.
factors, interferons, interleukins, and lymphokines. des--carboxy coagulation factors and coagulation
cytomegalovirus (CMV) Group of DNA viruses of the control proteins Coagulation cascade procoagulants II,
family Herpesviridae. CMV infection is asymptomatic in VII, IX, and X; and control proteins C, S, and Z that require
adults but can be transmitted to a fetus in utero to cause a vitamin Kcatalyzed -carboxylation of glutamic acid.
potentially fatal infection or reduced intelligence. CMV can During anticoagulant therapy with warfarin (Coumadin),
be transmitted by blood transfusions and is detected using which suppresses vitamin K, these des--carboxylated pro-
molecular diagnostic techniques. teins cannot participate in coagulation.
cytopenia Reduced cell count in one or more of any desquamation Shedding of epithelial elements, chiefly of
blood cell linered blood cells, white blood cells, or the skin, in scales or sheets.
platelets. Diamond-Blackfan syndrome (congenital pure red cell
cytosol Fluid portion of the cytoplasm, less granules and aplasia) Rare congenital anemia of unknown etiology,
organelles, as separated by ultracentrifugation. Also called evident in the first 3 months of life. Anemia is severe and
cell sap. erythropoietin resistant; the reticulocyte count does not rise.
cytotoxic Describes a compound or agent that destroys or Platelet and white blood cell counts are normal.
damages cells. diapedesis Outward passage of white blood cells through
dacryocyte (teardrop cell) Red blood cell with a single intact vessel walls.
pointed extension, resembling a teardrop. Dacryocytes are differential white blood cell count Review and tabulation
often seen in the myeloproliferative neoplasm termed of 100 to 200 white blood cells (WBCs) in a stained blood
myelofibrosis with myeloid metaplasia. film. The different types of WBCs are counted and reported
D-dimer (D:D, D-D) One of the fibrin degradation prod- as absolute counts or percentages of total WBCs. In auto-
ucts. D-dimer is composed of two fibrin D fragments cova- mated hematology analyzers, the differential WBC count is
lently joined by factor XIII. The D-dimer assay is used to accomplished using flow cytometry counting several thou-
rule out venous thromboembolic disease and disseminated sand WBCs.
intravascular coagulation, and may be used to monitor the dilute Russell viper venom time (DRVVT) Coagulation
efficacy and length of warfarin therapy. test performed like a prothrombin time assay. Russell viper
deep vein thrombosis (DVT, deep venous thrombo- venom activates the common coagulation pathway at the
sis) Pathologic formation of a clot in a deep leg vein such level of factor X. In the DRVVT assay, the reagent is diluted
as the femoral vein. A manifestation of venous thromboem- 1:500. DRVVT is prolonged by lupus anticoagulant, and the
bolic disease that is associated with a number of acquired test is used routinely in screening for this antibody.
or congenital thrombotic risk factors, deep vein thrombosis diploid Having two sets of chromosomes, as normally found
raises the risk of a pulmonary embolus. in nuclei of somatic cells. In humans, the normal diploid
11-dehydrothromboxane B2 (11-DHT) Measurable urine number is 46.
product of the platelet cyclooxygenase (eicosanoid) activa- direct antiglobulin test (DAT, Coombs test) Screening
tion pathway. Employed clinically to measure in vivo plate- procedure in which antihuman globulin is used to detect
let activation and aspirin resistance. antibodies and complement bound to red blood cells
delayed hemolytic transfusion reaction Hemolysis that in vivo.
occurs days or weeks after a blood transfusion, caused by direct thrombin inhibitors (DTIs) Class of intravenous
an anamnestic response to the transfused red blood cells in therapeutic anticoagulants including argatroban, lepirudin,
a patient alloimmunized from a previous pregnancy or dabigatran, and bivalirudin that suppress coagulation by
transfusion. directly neutralizing thrombin. DTIs are used in place of
check Quality control process in which a current analyte heparin when heparin-induced thrombocytopenia with
result is compared with the result for the same analyte from thrombosis is suspected. DTI therapy may be monitored
the previous specimen from the same patient. using the partial thromboplastin time.
-granule (-body, dense body, dense granule) Platelet disseminated intravascular coagulation (DIC) Uncon-
organelle that contains and secretes the small molecules trolled activation of thrombin and consumption of coagula-
adenosine diphosphate, adenosine triphosphate, serotonin, tion factors, platelets, and fibrinolytic proteins secondary to
calcium (Ca2+), and magnesium (Mg2+). There are two to many initiating events, including infection, inflammation,
seven -granules per platelet, and they are named by their shock, and trauma. Most commonly evidenced by diffuse
opaque appearance in transmission electron microscopy. mucocutaneous bleeding.
deoxyhemoglobin Hemoglobin that is not combined with Dhle bodies In Wright-stained peripheral blood films, gray
oxygen, formed when oxyhemoglobin releases its oxygen to to light blue round or oval inclusions composed of ribo-
the tissues. somal RNA found singly or in multiples near the inner
deoxyribonucleic acid (DNA) Double-stranded, helical membrane surface of granulocyte cytoplasm.
nucleic acid that carries genetic information. DNA is com- dominant Denoting an inherited allele whose trait is expressed
posed of nucleotide sequences with four repeating bases: whenever the gene is present, even in heterozygotes.
APPENDIX Glossary 823

Donath-Landsteiner (D-L) autoantibody IgG autoanti- edema Accumulation of excess serous fluid in a fluid com-
body with anti-P specificity that binds red blood cells at 18 partment or tissue.
C and causes hemolysis at 37 C. Donath-Landsteiner anti- effusion Seepage and accumulation of plasma-derived fluid
body causes hemolysis in patients with paroxysmal cold into a body cavity from blood vessels as a result of blood
hemoglobinuria. vessel damage or hydrostatic pressure.
Down syndrome Congenital group of physical, mental, and Ehrlichia Genus of small, spherical to ellipsoid, nonmotile
functional abnormalities including distinctive facial fea- gram-negative bacteria transmitted by ticks that cause
tures, congenital heart disease, muscular hypotonia, and ehrlichiosis. Symptoms of infection include chronic fatigue,
mental retardation, all associated with trisomy 21. Approxi- malaise, and fever.
mately 10% of infants with Down syndrome are born with electronic impedance Opposition to the flow of electrical
or develop transient myeloproliferative disease (TMD), current. The impedance principle of cell counting is based
which resembles chronic myelogenous leukemia but on the detection and measurement of changes in electrical
resolves spontaneously. Within a few weeks of TMD resolu- resistance produced by cells as they transverse a small aper-
tion, the child has a 10% to 30% chance of developing acute ture in a conducting solution.
myeloid or acute lymphoblastic leukemia. electrophoresis Separation and identification of proteins
drepanocyte (sickle cell) Abnormal crescent-shaped red and hemoglobin types based on their relative rates of migra-
blood cell containing hemoglobin S, characteristic of sickle tion through agarose or polyacrylamide gel in an applied
cell anemia. electrical field. The rate of migration is based on molecular
drug-induced hemolytic anemia Hemolytic anemia mass and net charge.
caused directly by a drug or secondary to an antibody- elliptocytes (ovalocytes) Oval red blood cells seen in the
mediated response stimulated by the drug. peripheral blood in the membrane disorder hereditary ellip-
dry tap Term used when an inadequate sample of bone tocytosis. Elliptocytosis may be asymptomatic or associated
marrow fluid is obtained during bone marrow aspiration. with mild anemia.
Dry taps occur when the marrow is packed, as in chronic elliptocytosis (ovalocytosis) Hematologic disorder char-
myelogenous leukemia, or when it is fibrotic, as in myelo- acterized by the presence of elliptocytes; often asymptom-
fibrosis with myeloid metaplasia. atic but may be associated with slight anemia.
duodenum Proximal portion of the small intestine adjacent Embden-Meyerhof pathway (EMP, glycolysis) A series of
to the stomach. enzymatically catalyzed reactions by which glucose and
dyscrasia Disorder of a hematologic cell line or lines. other sugars are metabolized to yield lactic acid (anaerobic
dyserythropoiesis Deranged erythropoiesis producing cells glycolysis) or pyruvic acid (aerobic glycolysis). Metabolism
with abnormal morphology; usually applied to congenital releases energy in the form of adenosine triphosphate.
dyserythropoietic anemia. embolism Pathologic event in which an embolus (foreign
dysfibrinogenemia Presence in the plasma of structurally object) travels through the bloodstream, becomes lodged in
abnormal fibrinogen; often a result of liver disease, occa- an artery, and obstructs blood flow. The embolus is often a
sionally congenital. blood clot, but it may be a fat globule, air bubble, piece of
dysmegakaryopoiesis Defective megakaryocytic production tissue, or clump of bacteria. Emboli occlude an artery and
and maturation characterized by cells with abnormal mor- cause tissue ischemia, necrosis, and loss of function. A pul-
phology and increased or decreased megakaryocyte counts. monary embolism is an embolus in a lung artery and is
dysmyelopoiesis Defective myelocytic production and mat- often fatal.
uration characterized by cells with abnormal morphology; endoplasmic reticulum (ER) Extensive network of
often applied to myelodysplastic syndromes. membrane-enclosed tubules in the cytoplasm of cells. The
dysplasia Abnormal growth pattern; for example, enlarged endoplasmic reticulum is rich in ribosomes that synthesize
skull in chronic anemia. Abnormal cervical epithelial cell proteins and provides a pathway for transport of protein
histopathologic features. through the cytoplasm.
dyspnea Difficult or painful breathing. endothelial cells Cell layer that lines the inner surface of all
ecchymosis Small hemorrhagic spot, larger than a petechia, blood vessels. Intact endothelial cells prevent thrombosis
in the skin or mucous membrane forming a rounded or because they present a smooth, nonactivating surface and
irregular blue or purplish patch. Also known as a bruise. secrete antiplatelet and anticoagulant substances. Injured
echinocyte (crenated red blood cell) Red blood cell endothelial cells promote clotting through exposure of
(RBC) with short, equally spaced, spiny projections; formed tissue factor and secretion of coagulation-promoting factors,
during cellular dehydration. Echinocytes are seen in associa- such as von Willebrand factor.
tion with uremia and pyruvate kinase deficiency and may enterocytes Epithelial cells that form the inner lining of the
be reversible. When echinocytes are unevenly distributed, intestine. Enterocytes absorb nutrients from the intestinal
they may be a drying artifact in blood film preparation. lumen and transport them to the portal circulation.
eclampsia Potentially life-threatening disorder during preg- eosinophil Granulocyte with large uniform cytoplasmic
nancy characterized by hypertension, generalized edema, granules that stain orange to pink with Wright stain. Gran-
proteinuria, and convulsions. ules usually do not obscure the segmented nucleus.
824 APPENDIX Glossary

eosinophilia Increase in the blood eosinophil count that is exon Portion of a DNA molecule that becomes translated to
associated with allergies, parasitic infections, or hemato- form a protein product. Exons and introns are transcribed
logic disorders. from DNA to heteronuclear RNA, where the introns are
epiphyses Ends of long bones that normally contain hema- excised to form messenger RNA.
topoietic tissue. extramedullary hematopoiesis (myeloid metaplasia)
epistaxis Hemorrhage from the nose; a nosebleed that Production of blood cells outside the bone marrow, such as
requires intervention. in the spleen, liver, or lymph nodes. Extramedullary hema-
Epstein-Barr virus (EBV) Herpesvirus that causes infectious topoiesis usually occurs in response to bone marrow fibrosis
mononucleosis and leads to the appearance of variant and loss of bone marrow hematopoiesis.
lymphocytes. extranodal Located outside a lymph node.
erythroblastosis fetalis (hemolytic disease of the fetus extravascular hemolysis Destruction of red blood cells
and newborn, HDFN) Alloimmune anemia caused by outside of a blood vessel, typically by splenic macrophage
maternal immunoglobulin G antibody that crosses the pla- phagocytosis.
centa and binds fetal red blood cell antigens inherited from extrinsic coagulation pathway Primary in vivo coagula-
the father; for instance, maternal anti-A with fetal A antigen. tion pathway. Exposure of tissue factor activates factor VII.
The disorder is characterized by hemolytic anemia, hyper- Factor VIIa activates factors IX and X, which triggers the
bilirubinemia, and extramedullary erythropoiesis. common pathway of coagulation and formation of fibrin.
erythrocyte (red blood cell, RBC) Nonnucleated bicon- exudate Fluid that has effused into a body cavity in associa-
cave disk-shaped peripheral blood cell containing hemoglo- tion with bacterial or viral infections, malignancy, pulmo-
bin. Its primary function is oxygen transport and delivery nary embolism, or systemic lupus erythematosus. An
to tissues. exudate is typically cloudy, because it contains cells and
erythrocyte sedimentation rate (ESR) Distance that red concentrated protein.
blood cells fall in a column of anticoagulated blood in factor assay Clot-based or chromogenic assay for specific
a specified time period. Elevated sedimentation rates are coagulation factor activity.
not specific for any disorder but indicate the presence of factor V Leiden mutation Substitution of arginine with
inflammation. glutamine at position 506 in the factor V protein. The resul-
erythrocytosis Increase in the red blood cell count in periph- tant factor V resists digestion by activated protein C. The
eral blood. factor V Leiden mutation results in increased thrombin pro-
erythroleukemia (Di Guglielmo disease, M6) Acute duction and is a thrombosis risk factor.
malignancy characterized by a proliferation of erythropoi- Fanconi anemia Autosomal recessive aplastic anemia mani-
etic precursors in bone marrow, erythroblasts with bizarre festing in childhood or early adult life. Characterized by
lobulated nuclei, and abnormal myeloblasts in peripheral mental retardation, absence of the thumb and radius, small
blood. stature, hypogonadism, and patchy brown discoloration of
erythron Total mass of red blood cells circulating in the the skin.
peripheral blood and their bone marrow precursors. favism Acute hemolytic anemia caused by ingestion of fava
erythropoiesis Bone marrow process of red blood cell beans or inhalation of the pollen of the plant. Usually
production. occurs in individuals with an inherited deficiency of
erythropoietin (EPO) Glycoprotein hormone synthesized glucose-6-phosphate dehydrogenase in red blood cells.
primarily in the kidneys and released into the bloodstream ferritin Iron-apoferritin complex, a major form in which iron
in response to anoxia. The hormone acts to stimulate and is stored in the body.
regulate the bone marrow production of red blood cells. ferrokinetics Study of iron metabolism, including the move-
essential thrombocythemia Myeloproliferative neoplasm ment of iron among the storage, transport, and functional
characterized by marked thrombocytosis and dysfunctional iron compartments.
platelets. Patients may experience bleeding or thrombosis. fibrin Fibrillar protein produced by the action of thrombin
etiology Causes or origin of a disease, for instance, genetic on fibrinogen in the clotting process. Fibrin is responsible
factors, infection, toxins, or trauma. Contrast with pathogen- for the semisolid character of a blood clot.
esis, which is the physiologic and biochemical mechanisms fibrin degradation products (FDPs, fibrin split prod-
by which a disease progresses. ucts, FSPs) Fibrin fragments X, Y, D, and E and D-dimer
euchromatin Portion of DNA that is active in gene expres- produced by the action of fibrin-bound plasmin during
sion and stains lightly with Wright stain. fibrinolysis.
euglobulin In the euglobulin lysis assay, the fraction of fibrinogen Plasma glycoprotein that is converted to fibrin by
treated plasma that contains fibrinogen, plasmin, plasmino- thrombin digestion.
gen and plasminogen activators, but lacks fibrinolytic fibrinolysis Continual process of digestion of fibrin by
inhibitors. bound plasmin that has been activated by bound plasmino-
exogenous Originating from outside the body or produced gen activator. Fibrinolysis is the normal mechanism for the
by external causes; used, for example, to describe a disease removal of fibrin clots. Fibrin is digested into fragments X,
caused by a bacterial or viral agent foreign to the body. Y, D, and E and D-dimer.
APPENDIX Glossary 825
fibroblast Connective tissue cell that differentiates into protein produced by myeloma tumor cells causes most
numerous cells which comprise the fibrous tissue of the gammopathies.
body, for instance, tissue in the walls of arteries. Gaucher disease Rare autosomal recessive disorder of fat
Fitzgerald factor (high-molecular-weight kininogen, metabolism caused by glucocerebrosidase deficiency and
HMWK) Member of the kinin inflammatory system that is characterized by histiocytic hyperplasia in the liver, spleen,
digested and activated by kallikrein to form bradykinin. lymph nodes, and bone marrow. The characteristic Gaucher
Fitzgerald factor is one of the in vitro contact activators of cells, which are lipid-filled macrophages whose cytoplasm
the coagulation system, which also include prekallikrein resembles crumpled tissue paper, are found on the Wright-
and factor XII. stained bone marrow aspirate smear.
Fletcher factor (prekallikrein, PK, pre-K) Member of the Gaussian distribution Frequency distribution that approxi-
kinin inflammatory system that forms active kallikrein upon mates the distribution of many random variables and is
digestion by kininogen. Fletcher factor helps to trigger in portrayed graphically as a symmetric bell-shaped curve. The
vitro coagulation contact activation. Fletcher factor defi- peak represents the mean of the distribution and the width
ciency prolongs the partial thromboplastin time but has no of the curve represents the dispersion or variability, which
clinical consequence. is generally expressed in terms of standard deviation from
flow cytometer Instrument in which cells suspended in fluid the mean. Also called a normal distribution.
flow one at a time through a focused beam of light. The gene Segment of a DNA molecule that contains all the infor-
light is scattered in patterns characteristic of the cells and mation required for synthesis of a protein, including both
their components. A sensor detecting the scattered or coding (exon) and noncoding (intron) sequences. Each
emitted light measures the size and molecular characteris- gene occupies a specific position (locus) on a particular
tics of individual cells. chromosome.
fluorescence in situ hybridization (FISH) Laboratory gene rearrangement Reorganization of the DNA sequences
technique in which fluorescence-labeled nucleic acid probes of a gene. Rearrangement of B-cell and T-cell genes produces
hybridize to selected DNA or RNA sequences in fixed tissue. an infinite variety of variable-region receptor and immuno-
FISH allows for the visual microscopic detection of specific globulin sequences. Clonal gene rearrangements occur in
polymorphisms or mutations such as BCR/ABL in cell or lymphoma and lymphocytic leukemia and are detected by
tissue specimens. flow cytometry to identify these diseases.
fluorophore Portion of a molecule that absorbs incident gestational age Age of the fetus, usually expressed as the
light and emits fluorescent light. Fluorophores are synthe- time elapsed from the first day of the mothers last men-
sized in molecules to provide measurable fluorescence in strual period.
laboratory assays. Glanzmann thrombasthenia (GT) Severe mucocutaneous
free erythrocyte protoporphyrin (FEP) Bib-iron porphy- bleeding disorder caused by one of a series of mutations in
rin precursor of heme that is present in low concentrations platelet glycoprotein (GP) IIb or IIIa. Normal GP IIb/IIIa
in normal red blood cells (RBCs). Elevated RBC concentra- recognizes and binds the arginine-glycine-aspartate peptide
tions indicate iron deficiency or impaired iron insertion. sequence receptor complex found in fibrinogen and von
French-American-British (FAB) classification Interna- Willebrand factor. Glanzmann thrombasthenia causes a
tional classification system for acute leukemias, myelopro- defect of fibrinogen-dependent platelet aggregation.
liferative neoplasms, and myelodysplastic syndromes globin Protein constituent of hemoglobin. Two identical
developed in the 1970s and 1980s. Still in use, although it pairs of globin chains bind four heme molecules to form
is being displaced by the World Health Organization hemoglobin.
classification. globulins Class of proteins that are insoluble in water or
fresh frozen plasma (FFP) Plasma that is separated by highly concentrated salt solutions but are soluble in mod-
centrifugation from whole-blood donations and frozen erately concentrated salt solutions. All plasma proteins are
within 8 hours of collection. FFP contains all of the plasma globulins except albumin and prealbumin.
procoagulants and control proteins, including the labile glossitis Inflammation of the tongue that causes it to be red
factors V and VIII. FFP is used for replacement therapy and smooth.
in acquired multiple-factor deficiencies, or in single-factor glucose-6-phosphate dehydrogenase (G6PD) First
deficiencies when factor concentrates are not available. enzyme of the glucose monophosphate shunt from the
G banding (GTG banding) In cytogenetic analysis, a pro- Embden-Meyerhoff pathway. G6PD catalyzes the oxidation
cedure in which metaphase chromosomes are treated with of glucose-6-phosphate to a lactone, converting the oxidized
trypsin, then stained with Giemsa dye. The areas rich in form of nicotinamide adenine dinucleotide phosphate
adenine-thymine, called G+, stain intensely, whereas the (NADP) to the reduced form (NADPH).
areas rich in guanine-cytosine (G) stain more lightly. The glucose-6-phosphate dehydrogenase deficiency X-linked
G+ bands correspond with Q bands in Q banding. Banding recessive deficiency of glucose-6-phosphate dehydrogenase
patterns are used for the identification of chromosomes. characterized by episodes of acute intravascular hemolysis
gammopathy Plasma protein imbalance caused by markedly under conditions of oxidative stress, including exposure to
increased concentrations of gamma globulin. A monoclonal oxidative drugs such as quinine.
826 APPENDIX Glossary

glycocalyx Glycoprotein and polysaccharide covering that projections resembling hairs. These cells are seen in hairy
surrounds many cells. The platelet glycocalyx is thicker cell leukemia.
than that of other cells and provides a procoagulant haploid Referring to the number of chromosomes found in
environment. sperm or ova, which is only one of each pair of chromo-
glycolysis (Embden-Meyerhof pathway, EMP) A series somes found in somatic (diploid) cells. In humans, the
of enzymatically catalyzed reactions by which glucose haploid number is 23.
and other sugars are metabolized to yield lactic acid haptoglobin Plasma protein that irreversibly binds free
(anaerobic glycolysis) or pyruvic acid (aerobic glycolysis). hemoglobin, forming a complex that is removed by macro-
Metabolism releases energy in the form of adenosine phages while conserving iron. Haptoglobin is decreased due
triphosphate. to consumption in intravascular hemolysis.
glycophorin Transverse red blood cell membrane protein Heinz bodies Round blue to purple inclusions in red blood
that carries several blood group antigens. cell visible when stained with new methylene blue dye.
glycoprotein Conjugated protein containing one or more Heinz bodies may be found in multiples and are composed
covalently linked carbohydrate residues. of precipitated hemoglobin in unstable hemoglobin
glycoprotein IIb/IIIa (GP IIb/IIIa) Pair of glycoproteins disorders.
that function as a receptor for the arginine-glycine-aspartate helmet cell Helmet-shaped schistocyte that may appear
sequence in fibrinogen and von Willebrand factor. The com- in microangiopathic hemolytic anemia (fragmented red
bination of fibrinogen and glycoprotein IIb/IIIa is essential blood cell).
to platelet aggregation. hemarthroses Chronic joint bleeds that cause inflammations
Golgi apparatus Rigid organelle comprised of numerous and immobilization; a symptom of severe hemophilia.
flattened sacs and associated vesicles. It is the location of hematemesis Vomiting of bright red blood.
the posttranslational modification and storage of glycopro- hematocrit (Hct, packed cell volume, PCV) Proportion
teins, lipoproteins, membrane-bound proteins, and lyso- of whole blood that consists of red blood cells, expressed
somal enzymes. as a percentage of the total blood volume.
gout Painful inflammation caused by excessive plasma uric hematoidin Golden yellow, brown, or red crystals that are
acid, which becomes deposited as monosodium urate chemically similar to bilirubin. Hematoidin crystal in a
monohydrate in joint capsules and adjacent tendons. tissue preparation indicates a hemorrhage site.
graft-versus-host disease (GVHD) Condition including hematology Clinical study of blood cells and blood-forming
tissue rejection that occurs when immunologically compe- tissues.
tent cells or their precursors are transplanted into an hematoma Localized collection of extravasated blood (blood
immunocompromised host who is not histocompatible that has escaped from vessel into tissue), usually clotted,
with the donor. The donor cells engraft and mount an in an organ space or tissue giving the appearance of a
immune response against the host. bruise.
granulocytes Class of red blood cells in peripheral blood hematopathology Study of the diseases of blood cells and
characterized by cytoplasmic granules; includes basophils, hematopoietic tissue.
eosinophils, and neutrophils. hematopoiesis Formation and development of blood cells.
granuloma Nodular, delimited aggregation of inflammatory Hematopoiesis occurs mostly in the bone marrow and
cells, macrophages, and macrophage-derived multinucleate peripheral lymphatic tissues.
giant cells surrounded by a rim of lymphocytes and fibro- hematopoietic microenvironment Matrix of bone marrow
blasts. Granulomas are major sites of cell-mediated immune stromal cells and tissue that supports hematopoiesis both
response to particulate antigens, and they may occur in structurally and through the production of cytokines and
many organs in chronic granulomatous disease. colony-stimulating factors.
GTG banding (G banding) In cytogenetic analysis, a pro- hematopoietic progenitor cell (HPC) Actively dividing
cedure in which metaphase chromosomes are treated with cell that is committed to a single blood cell lineage and is
trypsin, then stained with Giemsa dye. The areas rich in not capable of self-renewal. Therapeutic hematopoietic pro-
adenine-thymine, called G+, stain intensely, whereas the genitor cell products intended for transplantation provide
areas rich in guanine-cytosine (G) stain more lightly. The both hematopoietic stem cells and progenitor cells.
G+ bands correspond with Q bands in Q banding. Banding hematopoietic stem cell (HSC) Actively dividing cell that
patterns are used for the identification of chromosomes. is capable of self-renewal and of differentiation into any
guaiac test Test performed on stool emulsions to detect blood cell lineage.
hemoglobin as evidence of gastrointestinal bleeding. Small hematuria Abnormal presence of intact red blood cells in the
amounts of blood may be invisible; therefore it is known urine; indicative of kidney or urinary tract disease.
as a test for occult (hidden) blood. The peroxidase activity heme Pigmented iron-containing nonprotein part of the
of hemoglobin in blood reacts with guaiac to yield a blue hemoglobin molecule. There are four heme groups in a
color. hemoglobin molecule, each containing one ferrous ion in
hairy cells Lymphocytes seen in the peripheral blood and the center. Oxygen binds the ferrous ion and is transported
bone marrow characterized by delicate gray cytoplasm with from an area of high to low oxygen concentration.
APPENDIX Glossary 827
hemochromatosis Disease of iron metabolism that is char- by maternal IgG antibody that crosses the placenta and
acterized by excess deposition of iron in the tissues. The binds fetal red blood cell antigens inherited from the father:
disease may be inherited or may develop as a complication for instance, maternal anti-A with fetal A antigen. HDFN is
of a hemolytic anemia, such as sickle cell anemia. characterized by hemolytic anemia, hyperbilirubinemia,
hemoconcentration Elevation of the red blood cell, white and extramedullary erythropoiesis.
blood cell, and platelet counts resulting from a decrease in hemolytic uremic syndrome (HUS) Severe microangio-
plasma volume, for example, in dehydration. pathic hemolytic anemia that often follows infection of the
hemocytometer (hemacytometer, counting chamber) gastrointestinal tract by Escherichia coli serotype O157:H7,
Device used to perform visual blood and body fluid cell which produces an exotoxin. It is characterized by renal
counts, consisting of a microscopic slide with a depression failure, thrombocytopenia, the appearance of schistocytes
whose polished glass base is marked with grids and into on the peripheral blood film, and severe mucocutaneous
which a measured volume of a sample of diluted blood is hemorrhage.
placed and covered with a cover glass. The number of cells hemopexin Plasma glycoprotein produced in the liver.
in the squares is counted under a microscope and used as Hemopexin binds free plasma hemoglobin in the absence
a representative sample for calculating the unit volume. of haptoglobin.
hemodialysis Extracorporeal circulation process that substi- hemophilia Group of hereditary anatomic bleeding disor-
tutes for the kidneys and removes wastes from the blood. ders caused by a deficiency of a single coagulation factor.
Hemodialysis is used to treat patients with renal failure. The The two most common forms are hemophilia A and hemo-
patients blood is shunted from the body through a dialysis philia B, deficiencies of factors VIII and IX, respectively.
machine for diffusion and ultrafiltration and is then hemophilia A (classic hemophilia) Sex-linked recessive
returned to the patients circulation. anatomic bleeding disorder caused by a deficiency of coagu-
hemoglobin (Hb, HGB) Tetramer composed of two identi- lation factor VIII.
cal globin chains, each of which transports a heme mole- hemophilia B (Christmas disease) Sex-linked recessive
cule. Hemoglobin is the primary constituent of red blood anatomic bleeding disorder caused by a deficiency of coagu-
cell cytoplasm and transports molecular oxygen from the lation factor IX.
lungs to the tissues and returns carbon dioxide to the lungs. hemophilia C (Rosenthal syndrome) Autosomal ana-
hemoglobin C crystal Reddish hexagonal cytoplasmic red tomic bleeding disorder caused by a deficiency of coagula-
blood cell crystal described as a gold bar, or Washington tion factor XI.
monument. Typical of homozygous hemoglobin C disease, hemorrhage Acute severe blood loss requiring intervention
crystals form as deoxyhemoglobin C polymerizes. and transfusions.
hemoglobin electrophoresis Separation and identification hemorrhagic disease of the newborn Neonatal anatomic
of normal and abnormal hemoglobin types based on their bleeding caused by vitamin K deficiency.
relative rates of migration through agarose or polyacryl- hemosiderin Intracellular storage form of iron found pre-
amide gel in an applied electric field. The rate of migration dominantly in liver, spleen, and bone marrow cells. Hemo-
depends on molecular mass and net charge. Examples of siderin is a breakdown product of ferritin that appears in
hemoglobin types include Hb A, Hb C, Hb F, and Hb S. iron overload and hemochromatosis. Hemosiderin may be
hemoglobin SC crystal Irregular reddish cytoplasmic red detected microscopically using the Prussian blue iron stain.
blood cell crystal described as a glove or pistol. Typical hemosiderinuria Presence in the urine of hemosiderin,
of double heterozygous hemoglobin SC disease, the crystals which can be visualized using Prussian blue iron stain; most
form as deoxyhemoglobin S and C polymerize. often an indicator of hemolysis.
hemoglobinemia Presence of free hemoglobin in the blood hemosiderosis Increased tissue iron stores without associ-
plasma; indicative of intravascular hemolysis. ated tissue damage; may progress to hemochromatosis.
hemoglobinopathy Autosomal recessive anemia character- hemostasis Process by which a series of platelet, endothelial
ized by structural variations in globin genes that result in cell, and plasma enzyme systems prevent blood loss through
the formation of abnormal globin chains. Examples are clot formation and maintain blood vessel patency.
sickle cell anemia and hemoglobin C disease. heparin Naturally occurring mucopolysaccharide anticoagu-
hemoglobinuria Visible reddish free hemoglobin in the lant classified as a glycosaminoglycan. Heparin is produced
urine; indicative of intravascular hemolysis. by basophils and mast cells. Heparin extracted from porcine
hemolysis Disruption of red blood cell membrane integrity mucosa is used as a therapeutic anticoagulant.
that destroys the cell and releases hemoglobin. heparin-induced thrombocytopenia with thrombosis
hemolytic anemia Anemia characterized by a shortened red (HIT) Morbid, often fatal effect of unfractionated heparin
blood cell (RBC) life span and inability of the bone marrow therapy. Up to 5% of patients receiving unfractionated
to adequately compensate by increasing RBC synthesis. heparin therapy for more than 5 days develop an antibody
Hemolytic anemia may be caused by extracellular or intra- to the heparin-platelet factor 4 complex. The antibody and
cellular disorders. target antigen complex bind platelet Fc receptors and acti-
hemolytic disease of the fetus and newborn (HDFN, vate platelets, which causes a decrease in platelet count by
erythroblastosis fetalis) Alloimmune anemia caused greater than 40% and venous and arterial thrombosis.
828 APPENDIX Glossary

hepatitis Inflammation of the liver that damages hepatocytes histiocyte (macrophage) Mononuclear phagocyte found in
and releases bilirubin into the plasma. all tissues; part of the immune system.
hepatitis B virus (HBV) Causative agent of hepatitis B. The histochemical analysis Use of specialized stains to detect
virus is transmitted by contaminated blood or blood prod- enzymes and other chemicals in tissues.
ucts, by sexual contact with an infected person, or by the histocompatibility Quality or state of immunologic similar-
use of contaminated needles and instruments. Severe infec- ity that allows cells or tissues from one individual to be
tion may cause prolonged illness, destruction of liver cells, successfully transplanted into another individual. The
cirrhosis, increased risk of liver cancer, or death. degree of compatibility is controlled by white blood cell
hepatocyte Parenchymal liver cell. (WBC) surface markers of the human WBC antigen (HLA)
hepatomegaly Abnormal enlargement of the liver; usually a system of the major histocompatibility complex. Grafts
sign of disease. from donors of the immediate family are more compatible
hepatosplenomegaly Abnormal enlargement of the spleen than those from unrelated donors.
and liver. histogram Graph of a frequency distribution. In hematology,
hereditary elliptocytosis (ovalocytosis) Hereditary spec- a histogram is customarily a line graph generated by an
trin defect characterized by the presence of elliptocytes in automated analyzer that depicts the frequencies of platelet,
the peripheral blood; often asymptomatic but may be asso- white blood cell, or red blood cell volumes in a cell
ciated with slight anemia. population.
hereditary pyropoikilocytosis Rare hereditary defect of histology Science concerned with the microscopic identifica-
spectrin that causes severe hemolytic anemia beginning in tion of cells and tissues used to identify cellular and tissue
childhood and extreme poikilocytosis with red blood cell disease.
morphology resembling that seen in burn patients. histone Nuclear protein that complexes with DNA to form
hereditary spherocytosis Hereditary spectrin defect that chromatin. Histones provide for DNA folding and conden-
results in loss of red blood cell membrane and causes hemo- sation and help regulate replication and transcription.
lytic anemia characterized by numerous spherocytes on the Hodgkin lymphoma Solid tumor characterized by painless,
peripheral blood film. progressive hypertrophy of lymphoid tissue, first evident in
hereditary stomatocytosis Hereditary defect of the red cervical lymph nodes. Characterized by Reed-Sternberg cells
blood cell membrane resulting in a complex group of dis- and reactive white blood cells in lymphoid tissue and
eases in which the hemolysis is mild to severe and stomato- splenomegaly.
cytes are seen on the peripheral blood film. homocysteine Naturally occurring sulfur-containing amino
hereditary xerocytosis Hereditary defect of the red blood acid formed in the metabolism of dietary methionine.
cell membrane that results in hemolytic anemia with Homocysteine concentration in plasma depends upon
dehydrated red blood cells and the presence of stomato- adequate intake of protein, vitamin B6, vitamin B12, and
cytes, target cells, and macrocytes on the peripheral blood folate.
film. homocysteinemia Elevated plasma homocysteine, an inde-
heterochromatin Portion of DNA that is inactive during pendent risk factor for arterial thrombosis, typically caused
transcription to messenger RNA and stains deeply with by vitamin B6, vitamin B12, or folate deficiency.
Wright stain. homocysteinuria Presence of homocysteine crystals in urine
heterophile antibody Antibody that reacts with an antigen sediment; indicates an inherited error in an enzyme of the
from a species other than that of the antigen which stimu- methionine-homocysteine metabolic pathway.
lated its production. For example, patients with infectious homozygous Having two identical alleles at corresponding
mononucleosis caused by the Epstein-Barr virus produce an loci on homologous chromosomes. An individual who is
antibody to the virus but also produce heterophile antibod- homozygous for a trait has inherited one identical allele for
ies that react with sheep red blood cells. that trait from each parent. A person who is homozygous
heterozygous Having two different alleles at corresponding for a genetic disease caused by a pair of recessive alleles
loci on homologous chromosomes. An individual who is manifests the disorder.
heterozygous for a trait has inherited an allele for that trait Howell-Jolly (H-J) bodies Round blue to purple inclusions
from one parent and an alternative allele from the other in red blood cells (RBCs), one per RBC, visible on Wright-
parent. A person who is heterozygous for a genetic disease stained peripheral blood films. Howell-Jolly bodies are
will manifest the disorder if it is caused by a dominant allele composed of DNA and may indicate severe anemia or
but will remain asymptomatic if the disease is caused by a splenectomy.
recessive allele. human leukocyte antigens (HLA, major histocom
high-molecular-weight kininogen (HMWK, Fitzgerald patibility complex, MHC) Cell membrane glycoprotein
factor) Member of the kinin inflammatory system that is system that forms the basis for antigen presentation in cel-
digested and activated by kallikrein to form bradykinin. lular and humoral immunity and enables the immune
HMWK is one of the in vitro contact activators of the system to distinguish between self and nonself. HLA class I
coagulation system, which also include prekallikrein and molecules (HLA-A, -B, -C) are found on most nucleated cells
factor XII. and on platelets. Class II antigens (HLA-DP, -DQ, -DR) are
APPENDIX Glossary 829

expressed on B lymphocytes, monocytes, macrophages, and icterus (bilirubinemia, hyperbilirubinemia) Excess bili-
dendritic cells. The HLA system is assayed by flow cytometry rubin in plasma, which imparts a gold color to the plasma;
to assess transplant tissue compatibility. may indicate hemolytic anemia, liver disease, or bile duct
humoral immunity Immune response mediated by B lym- occlusion.
phocytes, which produce circulating antibodies (immuno- idiopathic Without a known cause.
globulins) in reaction to infectious organisms and other immediate transfusion reaction Hemolysis that begins
foreign antigens. within minutes or hours of a blood transfusion and is
hybridization In molecular biology, formation of a partially most commonly caused by an incompatibility of the ABO
or wholly complementary nucleic acid duplex by associa- system.
tion of single strands; used to detect and isolate specific immersion oil Optically clear oil placed in the space between
nucleic acid sequences. a microscopic specimen and the microscope objective lens.
hydrops fetalis Gross edema of the entire body of a fetus or Oil raises the refractive index, improving resolution.
newborn infant, associated with severe anemia and occur- immune hemolytic anemia Anemia resulting from short-
ring in hemolytic disease of the fetus and newborn. ened red blood cell (RBC) life span caused by antibodies to
hyperbilirubinemia (icterus, bilirubinemia) Excess bili- RBC membrane antigens or complement. The immuno-
rubin in the plasma, which imparts a gold color to the globulin- or complement-coated RBCs are cleared by splenic
plasma; may indicate hemolytic anemia, liver disease, or macrophages. Anemia results when the bone marrow fails
bile duct occlusion. to compensate for RBC consumption.
hypercellular bone marrow Bone marrow showing an immune thrombocytopenic purpura (ITP, idiopathic
abnormal increase in the concentration of nucleated hema- thrombocytopenic purpura, autoimmune thrombo-
topoietic cells; potentially associated with leukemia or cytopenic purpura, AITP) Mucocutaneous bleeding sec-
hemolytic anemia. ondary to thrombocytopenia caused by a platelet-specific
hypercoagulability (thrombophilia) Abnormally increased autoantibody that shortens platelet life span. Affects middle-
tendency to develop pathologic thromboses (clots) caused aged adults and is found more often in women than in men.
by a number of acquired and congenital factors. immunoblast Large mitotically active T or B lymphocyte
hyperplasia Abnormally increased number of cells per unit formed as a result of antigenic stimulation.
volume of tissue caused by increased cellular division or immunocompromised Unable to mount an adequate
abnormal retention. In bone marrow, hyperplasia is evident immune response due to disease or treatment with
in hypercellularity. Such hyperplasia may cause enlarged an immunosuppressive agent. An immunocompromised
joints or an enlarged forehead, called frontal bossing. patient is susceptible to bacterial and viral infections.
hypersegmented neutrophil Neutrophil with six or more immunocytochemical assays Laboratory tests in which
segments; often associated with megaloblastic anemia. antibodies labeled with chromophores or fluorophores
hypersplenism Increased hemolytic activity of the spleen hybridize specific cellular proteins or nucleic acids to
caused by splenomegaly, resulting in deficiency of periph- produce a measurable color or fluorescent reaction.
eral blood cells and compensatory hypercellularity of the immunoglobulin (antibody) Protein of the -globulin
bone marrow. fraction produced by B lymphocytes and plasma cells that
hypertension Persistently elevated blood pressure. recognizes and binds a specific protein antigen. Immuno-
hyperuricemia Excess of plasma uric acid or urates; some- globulins are the basis of humoral immunity.
times associated with gout. immunophenotyping Classification of white blood cells
hypocellular bone marrow Abnormal decrease in the and platelets by their membrane antigens. Synthetic anti-
number of nucleated hematopoietic cells present in the bodies, often monoclonal antibodies produced by hybrid-
bone marrow; may be associated with aplastic anemia or oma technology, are used to identify the antigens in flow
fibrosis. cytometry.
hypochromia Abnormal decrease in the hemoglobin content immunosuppression Abnormal state of the immune system
of red blood cells so that they appear pale when stained characterized by its inability to respond to antigenic stimuli.
with Wright stain. These cells are called hypochromic. Immunosuppressive drugs are used to reduce immune
hypodiploid Having fewer than the normal number of responses, particularly in tissue transplant therapy.
somatic cell chromosomes; for example, fewer than 46 chro- infectious mononucleosis Acute infection caused by the
mosomes in humans. Epstein-Barr virus, a herpesvirus. Characterized by fever, sore
hypoplasia Underdevelopment of an organ or tissue, usually throat, lymphadenopathy, variant lymphocytes, splenomeg-
resulting from a fewer-than-normal number of cells. A aly, hepatomegaly, abnormal liver function, and bruising.
hypoplastic bone marrow is one in which the distribution Laboratory tests used to identify the disease include blood
of nucleated hematopoietic cells is reduced, as in aplastic film review for variant lymphocytes, serologic mononucleo-
anemia or fibrosis. sis testing, and molecular identification of Epstein-Barr virus.
hypoxia Diminished availability of oxygen to body tissues, integrin Any of a family of cell-adhesion receptors that
usually secondary to decreased lung capacity or decreased mediate interactions between cells and between cells and
oxygen-carrying capacity of the blood. the extracellular matrix.
830 APPENDIX Glossary

interferon Natural glycoprotein produced by lymphocytes intron Nontranslated sequence in a gene. Introns are tran-
exposed to a virus or another foreign particle of nucleic acid. scribed to heteronuclear RNA and are excised during the
Interferon induces the production of translation inhibitory subsequent formation of messenger RNA.
protein (TIP) in noninfected cells. TIP blocks the translation inversion Structural chromosome alteration caused by breaks
of viral RNA and thus gives other cells protection against at two locations, reversal of direction of the detached
the original as well as other viruses. sequence, and reattachment. Inversions have little effect on
interleukin (IL) Any of a group of compounds that are syn- somatic DNA but cause loss of DNA information in
thesized by lymphocytes, macrophages, and matrix cells in offspring.
the marrow and interact with cells to initiate, stimulate, or iron deficiency anemia Microcytic, hypochromic anemia
influence the maturation of blood cells. caused by inadequate supplies of the iron needed to synthe-
international normalized ratio (INR) Index computed to size hemoglobin and characterized by pallor, fatigue, and
normalize prothrombin time (PT) results worldwide. The weakness. Often caused by low dietary iron intake or chronic
activity of a PT reagent (thromboplastin) is characterized by blood loss.
manufacturers using the international sensitivity index isoantibody (alloantibody) Antibody that is produced in
(ISI), which compares the reagent to the international refer- response to the presence of foreign antigens; for instance,
ence thromboplastin preparation provided by the World an antibody to a therapeutic coagulation factor that may
Health Organization. Local PTs are adjusted using the fol- render factor therapy ineffective.
lowing formula: INR = (PTpatient/PTmean)ISI, where PTpatient is jaundice Orange-yellow discoloration of the skin, mucous
the individual patients PT and PTmean is the mean of the membranes, and sclera. Caused by elevated plasma biliru-
range of normal PT values. bin, which signals hepatitis, hemolytic anemia, or common
international reference preparation (IRP) Human bile duct obstruction.
brainderived thromboplastin maintained by the World karyorrhexis Nuclear necrosis in which the nucleus ruptures
Health Organization. Thromboplastin manufacturers and chromatin disintegrates into formless granules.
worldwide compare their reagents sensitivity with that of karyotype Number, form, size, and arrangement of the chro-
this reference preparation using orthogonal regression to mosomes within the nucleus. In cytogenetic laboratory
generate an international sensitivity index (ISI) for their assays mitosis is halted in metaphase and the chromo-
products. somes are recorded in a photomicrograph to generate the
international sensitivity index (ISI) Index comparing the karyotype.
sensitivity of a given thromboplastin preparation with the kinin Any of a group of polypeptides that trigger inflamma-
sensitivity of the international reference preparation as tory activity such as contraction of smooth muscle, vascular
determined using orthogonal regression. The international permeability, and vasodilation. Examples of kinins are bra-
sensitivity index is used as an exponent in the equation for dykinin and kallidin.
calculating the international normalized ratio. kininogen Either of two plasma 2-globulins that are kinin
intracranial hemorrhage (ICH, hemorrhagic stroke) precursors, called high-molecular-weight kininogen and low-
Bleeding into the brain causing tissue death. Fifteen percent molecular-weight kininogen.
of strokes are caused by intracranial hemorrhage; the Kleihauer-Betke stain (acid elution slide test) Test for
remainder are caused by vascular occlusion (ischemia). detecting fetal red blood cells (RBCs) in the maternal circu-
intramedullary hematopoiesis Formation and develop- lation. Blood films are immersed in an acid buffer, which
ment of blood cells within the marrow cavity of a bone. causes adult hemoglobin (Hb A) to be eluted from RBCs.
intramuscular (IM) Injected into muscle tissue. The film is stained, and RBCs that have fetal hemoglobin
intravascular hemolysis Red blood cell destruction that (Hb F) take up the stain.
occurs within the blood vessels at a rate exceeding splenic Khler illumination Light microscope illumination system
macrophage clearance capacity, releasing hemoglobin that consists of a substage light source, field diaphragm,
into the plasma. Seen in acute hemolytic episodes such condenser, and aperture diaphragm. These are adjusted in
as those associated with transfusion reaction, glucose-6- sequence to optimize specimen illumination, resolution,
phosphate dehydrogenase deficiency, and sickle cell anemia contrast, and depth of field.
crisis. koilonychia Dystrophy of the fingernails in which they
intrinsic coagulation pathway Sequence of serine protease become thin, ridged, and concave. Associated with iron defi-
reactions leading to fibrin formation, beginning with the in ciency anemia.
vitro contact activation of factor XII, followed by the sequen- Kupffer cells Fixed, highly phagocytic macrophages that line
tial activation of factors XI and IX, and resulting in the the liver sinusoids. Kupffer cells function like splenic mac-
activation of factor X, which initiates the common pathway rophages (littoral cells) to clear senescent red blood cells,
of coagulation. immune complexes, and foreign materials.
intrinsic factor (IF) Glycoprotein secreted by parietal cells lactate dehydrogenase (LD) Enzyme that catalyzes the
of the gastric mucosa that is essential for the intestinal reversible conversion of lactate to pyruvate. It is widespread
absorption of vitamin B12. A deficiency of, or antibody to, in tissues and is particularly abundant in the kidneys,
intrinsic factor results in B12 deficiency. skeletal muscle, liver, red blood cells, and myocardium.
APPENDIX Glossary 831

The lactate dehydrogenase assay may be used to detect and low-molecular-weight heparin (LMWH) Heparin with
monitor cell necrosis in these organs, for instance, acute an average molecular weight of 5000 to 8000 produced by
myocardial infarction. enzymatic or chemical digestion of unfractionated heparin.
Langerhans cell Immature dendritic macrophage (histio- LMWH is used for prophylaxis, and treatment monitoring
cyte) with an irregular nucleus and distinctive racquet- is required only in conditions of fluid imbalance, such as
shaped cytoplasmic granules called Birbeck bodies normally obesity, renal disease, and pregnancy. LMWH therapy is
found in the epidermis, oral and vaginal mucosa, and lungs. monitored using the chromogenic antifactor Xa heparin
Langerhans cells have been identified as the cell of origin assay.
for a nonproliferating epidermal neoplasm known as lupus anticoagulant (LA, LAC) Autoantibody to phospho-
Langerhans cell histiocytosis. lipid-binding proteins such as 2-glycoprotein I, annexin,
leptocyte Abnormal mature red blood cell that is thin and and prothrombin. May be present as a primary condition
flat with hemoglobin at the periphery and increased central or secondary to a collagen disorder such as systemic lupus
pallor. erythematosus, Sjgren syndrome, or rheumatoid arthritis.
leukemia Group of malignant neoplasms of hematopoietic Chronic lupus anticoagulant is associated with venous and
tissues characterized by diffuse replacement of bone marrow arterial thrombosis and spontaneous abortion.
or lymph nodes with proliferating white blood cell (WBC) lymphadenopathy Any disorder characterized by a localized
precursors. WBC counts become elevated, and immature or generalized enlargement of the lymph nodes or lymph
and variant forms appear in the peripheral blood. Leukemia vessels.
may be chronic or acute, myeloid or erythroid. lymphoblast Immature cell found in the bone marrow and
leukemoid reaction (LR) Clinical syndrome resembling lymph nodes, but not normally in the peripheral blood;
leukemia in which the white blood cell count is elevated to the most primitive, morphologically recognizable precursor
greater than 25,000/mcL in response to an allergen, inflam- in the lymphocytic series, which develops into the
matory disease, infection, poison, hemorrhage, burn, or prolymphocyte.
severe physical stress. Leukemoid reaction usually involves lymphocyte Family of mononuclear, nonphagocytic white
granulocytes and is distinguished from chronic myeloge- blood cells found in the blood, lymph, and lymphoid
nous leukemia by the use of the leukocyte alkaline phos- tissues. Lymphocytes are categorized as B and T lympho-
phatase staining of neutrophils. cytes and natural killer cells. They are responsible for
leukocyte One of the formed elements of the blood. The five humoral and cellular immunity and tumor surveillance.
families of WBCs are lymphocytes, monocytes, neutrophils, lymphocytopenia (lymphopenia) Abnormally reduced
basophils, and eosinophils. WBCs function as phagocytes lymphocyte count in peripheral blood.
of bacteria, fungi, and viruses; detoxifiers of toxic proteins lymphocytosis Abnormally increased lymphocyte count in
that may be produced by allergic reactions and cellular peripheral blood.
injury; and immune system cells. lymphoid Resembling or pertaining to lymph or tissue and
leukocytosis Abnormally elevated white blood cell count in cells of the lymphoid system.
peripheral blood. lymphokines Biologic response mediators released by both
leukoerythroblastic Characterized by the presence of imma- B and T lymphocytes.
ture red blood cells and granulocytes in the peripheral lymphoma Solid tumor neoplasm of lymphoid tissue catego-
blood and bone marrow. rized as Hodgkin or non-Hodgkin lymphoma and defined
leukopenia Abnormal decrease in the white blood cell count by lymphocyte morphology and the histologic features of
in peripheral blood. the lymph nodes.
leukopoiesis Process by which white blood cells form and lymphopoiesis Formation and production of lymphocytes,
develop in the bone marrow and lymph nodes. predominantly in the lymph nodes.
Levey-Jennings chart Quality control chart used to plot lymphoproliferative Pertaining to the proliferation of lym-
periodic test results for control specimens. The chart indi- phoid cells resulting in abnormally increased lymphocyte
cates the mean and the 1, 2, and 3 standard deviation counts in peripheral blood, indicating a reactive or neoplas-
intervals on both sides of the mean. Deviation from this tic condition.
standard distribution indicates the occurrence of a system- lysosomes Membrane-bound sacs of varying size distributed
atic analytical error. randomly in the cytoplasm of granulocytes and platelets.
ligand Molecule, ion, or group bound to the central atom of Lysosomes contain hydrolytic enzymes that kill ingested
a chemical compound; for example, the oxygen molecule in bacteria and digest bacteria and other foreign materials.
hemoglobin, which is bound to the central iron atom. Also, macrocyte Red blood cell with an abnormally large diameter
a molecule that binds to another molecule, used especially seen on a peripheral blood film. Associated with folate and
to refer to a small molecule that specifically binds to a larger vitamin B12 deficiency or refractory anemia.
molecule. macroglobulin High-molecular-weight plasma globulin,
littoral cells Fixed, highly phagocytic macrophages that line such as 2-macroglobulin or an immunoglobulin of the
the sinusoid of the spleen. Littoral cells clear senescent red M isotype. Abnormal monoclonal IgM proteins seen in
blood cells, immune complexes, and foreign materials. Waldenstrm macroglobulinemia.
832 APPENDIX Glossary

macrophage (histiocyte) Mononuclear phagocyte found in megakaryopoiesis (megakaryocytopoiesis) Production


all tissues; part of the immune system. and development of megakaryocytes, the precursors to
major histocompatibility complex (MHC, human platelets, in the bone marrow.
leukocyte antigen, HLA) Cell membrane glycoprotein megaloblast Abnormally large, nucleated, immature precur-
system that forms the basis for antigen presentation in sor of the erythrocytic series; an abnormal counterpart to
cellular and humoral immunity and enables the immune the pronormoblast. Not only does it have a larger diameter,
system to distinguish between self and nonself. MHC class but the nucleus appears more immature than the cyto-
I molecules (HLA-A, -B, -C) are found on most nucleated plasm. Megaloblasts give rise to macrocytic red blood cells
cells and on platelets. Class II antigens (HLA-DP, -DQ, -DR) and are associated with megaloblastic anemia, usually
are expressed on B lymphocytes, monocytes, macrophages, caused by folate or vitamin B12 deficiency.
and dendritic cells. The HLA system is assayed by flow menorrhagia Abnormally heavy or prolonged menstrual
cytometry to assess transplant tissue compatibility. periods.
malaise Vague feeling of discomfort and fatigue, often associ- metacentric Describes a mitotic chromosome having the
ated with cancer or anemia. centromere at the center, with one arm equal in length to
malaria Infectious disease caused by one or more of at least the other.
four species of the protozoan genus Plasmodium. Malaria is metamyelocyte Stage in the development of the granulocyte
transmitted from human to human by a bite from an series, located between the myelocyte stage and the band
infected Anopheles mosquito. stage. Characterized by mature, granulated cytoplasm and a
malignant Describes a cancerous disease that threatens life bean-shaped nucleus.
through its ability to metastasize. metaphase Second phase of mitosis in which the chromo-
marker chromosome In cytogenetic analysis, a chromo- somes are aligned at the equatorial plate. Chromosomes at
some of abnormal size or shape that is an early indicator of metaphase are maximally contracted and are most easily
neoplastic disease, for instance, the Philadelphia chromo- identified in cytogenetic analysis.
some in chronic myelogenous leukemia. metaplasia Conversion of normal tissue cells into another,
mast cell Connective tissue cell that has large basophilic less differentiated cell type in response to chronic stress or
granules containing heparin, serotonin, bradykinin, and injury. For instance, myeloid metaplasia describes hematopoi-
histamine. These substances are released from the mast cell esis in the spleen or liver.
in response to immunoglobulin E stimulation. metarubricyte (orthochromic normoblast) Fourth stage
May-Hegglin anomaly Rare autosomal dominant disorder of bone marrow erythropoiesis and the last in which the
characterized by thrombocytopenia and granulocytes that cell retains a nucleus. The nucleus is fully condensed with
contain cytoplasmic inclusions similar to Dhle bodies. no parachromatin, and the cytoplasm is 85% hemoglo-
mean Value that is derived by dividing the total of a set of binized and gray-red. In hemolytic anemia, when an ortho-
values by the number of items in the set; the arithmetic chromic normoblast appears in the peripheral blood, it is
average. called a nucleated red blood cell.
mean corpuscular hemoglobin (MCH) Average red metastasis Extension or spread of tumor cells to distant parts
blood cell (RBC) mass in picograms computed from the of the body, usually through the lymphatics or blood
RBC count and hemoglobin level. vessels.
mean corpuscular hemoglobin concentration (MCHC) methemoglobin Abnormal form of hemoglobin in which
Average relative hemoglobin per red blood cell (RBC), the ferrous ion has become oxidized to the ferric state. Met-
expressed in percent, computed from the hemoglobin and hemoglobin cannot carry oxygen.
hematocrit. Relates to Wright-stained RBC color intensity. microangiopathic hemolytic anemia (MAHA) Condi-
mean corpuscular volume (MCV) Average red blood cell tion in which narrowing or obstruction of small blood
(RBC) volume in femtoliters computed from the RBC count vessels by fibrin results in distortion and fragmentation of
and hematocrit or directly measured by an impedance or red blood cells, hemolysis, and anemia. This causes the
light-activated particle counter. Relates to Wright-stained appearance of schistocytes on a Wright-stained blood film.
RBC diameter. microcyte Small red blood cell (RBC) with reduced mean
megakaryoblast Least differentiated visually identifiable corpuscular volume and reduced RBC diameter on Wright-
megakaryocyte precursor in a Wright-stained bone marrow stained peripheral blood film. Microcytes are often associ-
aspirate smear. The megakaryoblast cannot be distinguished ated with iron deficiency anemia and thalassemia.
visually from the myeloblast but is identified using special microfilament Intracellular protein that supports the cyto-
immunochemical markers. skeleton and assists with cell motility.
megakaryocyte Largest cell in the bone marrow, measuring micrometer reticule Scaled optical disc installed in a micro-
30- to 50-m and having a multilobed nucleus. Its cyto- scope ocular lens; calibrated in micrometers to measure cell
plasm is composed of platelets, which are released to the diameter during peripheral blood film examination.
blood through the extension of proplatelet processes. Mega- microtubules Tubulin protein channels that maintain cel-
karyocytes are identified and enumerated microscopically at lular shape, contribute to motility, and make up mitotic
low (10) power. spindle fibers and centrioles.
APPENDIX Glossary 833
mitochondria Round or oval structures distributed ran- neutrophils. Lymphoid and erythroid cell lines are excluded,
domly in the cytoplasm of a cell. Mitochondria provide the and most morphologists also exclude the monocytic and
cells aerobic energy system by producing adenosine megakaryocytic cell lines.
triphosphate. myeloid-to-erythroid (M:E) ratio Proportion of myeloid
mitosis Ordinary process of somatic cell division resulting in cells to nucleated erythroid precursors in bone marrow aspi-
the production of two daughter cells that have identical rate. The myeloid-to-erythroid ratio is used to evaluate
diploid complements of chromosomes. hematologic cell production. Excluded from the myeloid
monoblast Most undifferentiated morphologically identifi- cell count are monocytic and lymphoid precursors and
able precursor of the bone marrow monocytic series; devel- plasma cells.
ops into the promonocyte. myelokathexis Inherited severe neutropenia and lymphope-
monoclonal Pertaining to or designating a group of identical nia characterized by hypersegmented neutrophils and
cells or organisms derived from a single cell or organism. myeloid hyperplasia. Part of WHIM syndrome (warts, hypo-
Also used to describe products from a clone of cells, such gammaglobulinemia, infections, and myelokathexis).
as monoclonal antibodies. myeloperoxidase (MPO) Enzyme that occurs in primary
monocyte Mononuclear phagocytic white blood cell having granules of promyelocytes, myelocytes, and neutrophils and
a round to horseshoe-shaped nucleus with abundant gray- exhibits bactericidal, fungicidal, and virucidal properties.
blue cytoplasm filled with fine reddish granules. Circulating Cytochemical stains that detect myeloperoxidase are used
precursor to the macrophage, the primary phagocytic cell of to identify myeloid precursors in acute leukemia.
most tissues. myeloproliferative neoplasms (MPN, myeloprolifera-
monocytopenia Abnormally low monocyte count in periph- tive disorders, MPD) Group of neoplasms characterized
eral blood. by proliferation of myeloid tissue and elevations in one or
monocytosis Abnormally elevated monocyte count in more myeloid cell types in the peripheral blood. Myelopro-
peripheral blood; may indicate infection or a neoplasm. liferative neoplasms include myelofibrosis with myeloid
mononuclear Having only one nucleus. Used to describe metaplasia, essential thrombocythemia, polycythemia vera,
cells such as monocytes or lymphocytes as distinct from and chronic myelogenous leukemia.
neutrophils, which have nuclei that appear to be multiple myoglobin Monomeric heme-containing protein in muscle.
and hence are called segmented or polymorphonuclear. Combines with oxygen released by red blood cells, stores it,
Mott cell Plasma cell containing colorless cytoplasmic inclu- and transports it to the mitochondria of muscle cells, where
sions of immunoglobulin called Russell bodies that appear it generates energy.
similar to vacuoles. necrosis Localized tissue death that occurs in groups of cells
multiple myeloma Malignant neoplasm in which plasma in response to disease or injury.
cells proliferate in the bone marrow, destroying bone and neonatal Pertaining to the first 28 days after birth.
resulting in pain, fractures, and excess production of a neoplasm Any abnormal growth of new tissue; can be malig-
monoclonal plasma immunoglobin. nant or benign. The term is usually applied to cancerous
mutation Permanent transmissible change in a single gene; cells.
includes substitution, loss, or gain of a base pair. Mutations neuropathy Any disorder affecting the nervous system.
cause the production of abnormal proteins or the loss of a neutropenia Abnormally reduced neutrophil count in
protein. peripheral blood. Often associated with chemotherapy, it
myelo- Prefix relating to the bone marrow or spinal cord and exposes the patient to the risk of infection.
used to identify granulocytic precursors of neutrophils. neutrophil Mature segmented (polymorphonuclear) white
myeloblast Least differentiated morphologically identifiable blood cell with fine pink-staining cytoplasmic granules in a
bone marrow precursor of the granulocytic series; develops Wright-stained peripheral blood film. Neutrophils ingest
into the promyelocyte. The appearance of myeloblasts in bacteria and cellular debris.
peripheral blood signals acute leukemia. neutrophilia Abnormally elevated neutrophil count in
myelocyte Third stage of bone marrow granulocytic series peripheral blood; often indicates bacterial infection and
differentiation, intermediate in development between a may be associated with chronic or acute leukemia.
promyelocyte and a metamyelocyte. In this stage, differen- nondisjunction Faulty distribution of chromosomal ele-
tiation of cytoplasmic granules has begun, so myelocytes ments during mitosis or meiosis.
may be basophilic, eosinophilic, or neutrophilic. non-Hodgkin lymphoma (NHL) Solid tumors of lym-
myelodysplastic syndromes (MDSs) Group of acquired phoid tissue classified by histologic features and lympho-
clonal hematologic disorders characterized by progressive cytic morphology. Should be distinguished from Hodgkin
peripheral blood cytopenias that reflect defects in erythroid, lymphoma, which is caused by proliferation of Reed-
myeloid, or megakaryocytic maturation. Sternberg cells and accumulation of reactive peripheral
myelofibrosis Replacement of bone marrow with fibrous blood cells.
connective tissue. nonsteroidal antiinflammatory drugs (NSAIDs) Anti-
myeloid General term used to denote granulocytic cells and inflammatory, analgesic, and antiplatelet drugs, other
their precursors, including basophils, eosinophils, and than steroids. NSAIDs include aspirin, which acetylates
834 APPENDIX Glossary

cyclooxygenase and reduces platelet activation, as well as and gray-red. In hemolytic anemia, when the orthochromic
naproxen, acetaminophen, and ibuprofen. normoblast appears in the peripheral blood, it is called a
normochromic Describes a Wright-stained red blood cell nucleated red blood cell.
with normal color and normal hemoglobin content. osmotic fragility test Assay in which whole blood is pipet-
normocyte Normal, mature red blood cell of average volume ted to each of a series of saline solutions of graduated
and color. concentration. The series begins with water and increases in
nucleated red blood cell (NRBC) Red blood cell (RBC) concentration to normal (0.85%) saline. The osmotic fragil-
in peripheral blood that possesses a nucleus; often an ity test is used to detect spherocytosis, because spherocytes
orthochromic normoblast (metarubricyte). Nucleated RBCs rupture in saline concentrations near the normal level. It
falsely raise automated white blood cell (WBC) counts, may also detect target cells, which, owing to their reduced
which requires a manual WBC count correction. hemoglobin content, are able to withstand osmotic stress
nucleolus Round or irregular pocket of messenger and ribo- and rupture only at very dilute saline concentrations.
somal RNA in the nucleus. Nucleoli are observed by mor- osteoblast Bone-forming cell.
phologists and are used to distinguish cell differentiation osteoclast Large multinuclear cell associated with the absorp-
stages. tion and removal of bone. May be confused with a
nucleoside Glycoside of purine and pyrimidine bases. In megakaryocyte.
RNA and DNA, the glycosides are the pentoses ribose and oval macrocyte Oval red blood cell with an increased diam-
deoxyribose. eter seen in peripheral blood. Characteristic of megaloblas-
nucleotide Phosphoric ester of a nucleoside. Allows for the tic anemia.
formation of phosphodiesterase bridges between nucleo- ovalocyte (elliptocyte) Oval red blood cell seen in periph-
tides to form RNA and DNA. eral blood in the membrane disorder hereditary elliptocy-
nucleus Cellular organelle containing DNA and RNA. Site of tosis. Elliptocytosis may be asymptomatic or associated with
genetic replication. mild anemia.
nucleus-to-cytoplasm (N:C) ratio Estimated volume of a oxygen affinity Ability of hemoglobin to bind oxygen
Wright-stained nucleus in comparison to the volume of the molecules.
cytoplasm. The nucleus-to-cytoplasm ratio is used to dif- oxygen dissociation curve Graphic expression of the affin-
ferentiate cell developmental stages. ity between oxygen and hemoglobin or the concentration
numeric aperture (NA) Number stamped on the barrel of of oxygen bound at equilibrium to the hemoglobin in
a microscope lens designating the quality of the microscope blood as a function of oxygen pressure.
objective. The higher the number, the greater the lenss oxyhemoglobin Hemoglobin that contains bound oxygen.
resolution. P arm Smaller (petite) arm of a chromosome.
objective Microscope lens closest to the specimen. Most clini- packed cell volume (PCV, hematocrit, Hct) Proportion
cal grade microscopes provide 10 dry, 50 oil immersion, of whole blood that consists of red blood cells, expressed
and 100 oil immersion objectives. as a percentage of the total blood volume.
ocular Microscope lens, usually 10, nearest the eye. pallor Unnatural paleness or absence of color from the skin.
oncogene Gene capable, under certain conditions, of causing pancytopenia Marked reduction in the count of red blood
the initial and continuing conversion of normal cells into cells, white blood cells, and platelets in peripheral blood.
cancer cells. A mutated proto-oncogene. Pappenheimer bodies (siderotic granules) Red blood
opsonization Process by which an antibody or complement cell inclusions composed of ferric iron. On Prussian blue
attaches itself to foreign material (bacteria), triggering or iron stain preparations, they appear as multiple dark blue
enhancing phagocytosis by white blood cells. irregular granules at the periphery. On Wright stain prepara-
optical scatter Scattering of light caused by the interaction tions they appear as pale blue clusters.
of absorption, diffraction, refraction, and reflection. Hema- parachromatin Pale-staining portion of the nucleus, roughly
tology analyzers that use both laser and nonlaser light apply equivalent to euchromatin.
the principle of light scatter to perform cell counting and parenteral Pertaining to administration of drugs or other
differentiation. The angle of light scatter correlates with compounds by means other than oral administration;
various cell features such as volume, density, and cellular includes intramuscular and intravenous administration.
complexity. paroxysmal cold hemoglobinuria (PCH) Rare acquired
oral anticoagulant therapy (OAT) Anticoagulant therapy autoimmune hemolytic anemia in which the Donath-
using warfarin (Coumadin), which functions as a vitamin Landsteiner autoantibody binds red blood cells during
K antagonist to prevent -carboxylation of the glutamic acid exposure to cold, producing acute hemolysis and hematuria
units in coagulation factors II (prothrombin), VII, IX, and upon warming.
X and thus slows thrombin production. paroxysmal nocturnal hemoglobinuria (PNH) Acquired
orthochromic normoblast (metarubricyte) Fourth stage hemolytic anemia due to a stem cell clonal mutation
of bone marrow erythropoiesis and the last in which the that causes the cell to lack glycosylphosphatidylinositol-
cell retains a nucleus. The nucleus is fully condensed with anchored proteins, including decay-accelerating factor and
no parachromatin, the cytoplasm is 85% hemoglobinized CD59, two proteins that normally protect the red blood cell
APPENDIX Glossary 835

(RBC) from complement activation and hemolysis. RBCs phlebotomy Use of a hypodermic needle to puncture a vein
therefore have an increased susceptibility to complement, and collect blood.
which results in intravascular hemolysis and hemoglobin- phytohemagglutinin (PHA) Lectin extracted from the red
uria that occur in irregular episodes, especially during sleep. kidney bean that causes red blood cell agglutination by
partial thromboplastin time, (PTT, activated partial binding to N-acetyl--glucosamine. Also a mitogen that
thromboplastin time, APTT) Clot-based screening induces T-lymphocyte proliferation in culture.
test for intrinsic coagulation, which is prolonged in defi- pinocytosis Cellular ingestion of small particles or liquids.
ciencies of prekallikrein, high-molecular-weight kininogen, pitting Removal by the spleen of material from within red
and factors XII, XI, IX, VIII, X, V, II, and I. Calcium chloride, blood cells (RBCs) without damage to the RBCs; for
phospholipid, and activator are added to patient plasma. example, removal of nuclei or Howell-Jolly bodies.
The interval from the addition of reagent to clot formation plasma Fluid portion of the blood in which the formed ele-
is recorded. The test is used to monitor unfractionated ments (white blood cells, red blood cells, and platelets) are
heparin therapy, to screen for intrinsic pathway deficiencies, suspended.
and to screen for lupus anticoagulant. plasma cell Fully differentiated B lymphocyte found in the
pathogenesis Chemical and biologic events in cells and bone marrow and lymphoid tissue, and occasionally in
tissue by which disease occurs and progresses. peripheral blood. It contains an eccentric nucleus with
pathognomonic Specifically characteristic of a given disease; deeply staining chromatin and abundant dark blue cyto-
indicating a sign or symptom from which a diagnosis can plasm. The Golgi apparatus produces a perinuclear halo due
be made. to its high lipid content. Plasma cells secrete antibody in
Pelger-Hut anomaly Autosomal dominant asymptomatic the humoral immune response.
anomaly of neutrophil nuclei, which fail to segment and plasma frozen within 24 hours (FP24) Plasma that is
appear dumbbell shaped or peanut shaped (pince-nez separated from whole-blood donations and frozen within
nuclei). Pelgeroid nuclei are more common and resemble 24 hours of collection. FP24 contains all of the plasma
the nuclei of Pelger-Hut anomaly, but may indicate myelo- procoagulants and control proteins, including adequate
dysplasia or may appear during chemotherapy. levels of the labile factors V and VIII. FP24 is used for
percutaneous coronary intervention (PCI) Any catheter- replacement therapy in acquired multiple-factor deficiencies
based technique for the management of coronary artery or in single-factor deficiencies when factor concentrates are
occlusion, such as angiography, angioplasty, stent place- not available.
ment within a coronary artery, or cardiac catheterization. plasmin Active form of plasminogen. Plasmin binds fibrin
peripheral artery occlusion (PAO) Blockage of a noncar- and, when activated by tissue plasminogen activator, digests
diovascular, noncerebral arteryfor example, a carotid or fibrin to form fibrin degradation products in the fibrinolytic
brachial arteryby thrombus formation. process.
pernicious anemia Progressive autoimmune disorder that plasminogen Inactive (zymogen) plasma precursor of
results in megaloblastic macrocytic anemia due to a lack of, plasmin.
or antibodies to, parietal cells or intrinsic factor essential for plasminogen activator Substance that cleaves plasminogen
the absorption of vitamin B12. and converts it into plasmin; includes urokinase, which is
personal protective equipment (PPE) Clothing that is secreted by renal endothelial cells, and tissue plasminogen
used to prevent blood or other potentially infectious bio- activator, which is secreted by all endothelia. Synthetic plas-
logic substances from contacting clothing, eyes, mouth, or minogen activators are used therapeutically to clear coro-
mucous membranes. Includes, for example, gloves, fluid- nary artery clots after acute myocardial infarction.
impermeable laboratory coats, and eye protection. plasminogen activator inhibitor 1 (PAI-1) Endothelial
petechiae Pinpoint purple or red spots on the skin or mucous cell inhibitor of tissue plasminogen activator; controls
membranes indicating small bleeds within the dermal fibrinolysis.
or submucosal layers. May indicate a systemic bleeding platelet (thrombocyte) Smallest of the formed elements in
disorder. blood; a disk-shaped, 2- to 4-m nonnucleated cell formed
phagocyte Cell that is able to surround, engulf, and digest in the bone marrow by fragmentation of megakaryocytes.
microorganisms and cellular debris. Macrophages and Platelets trigger and control blood coagulation.
neutrophils are phagocytes. platelet adhesion Platelet attachment to subendothelial col-
phagocytosis Ingestion of large particles or live microorgan- lagen, part of a sequential mechanism leading to the initia-
isms into a cell. tion and formation of a thrombus or hemostatic plug.
phagosome Membrane-bound cytoplasmic vesicle in a Platelet adhesion requires von Willebrand factor and plate-
phagocyte containing the phagocytosed material. let receptor glycoprotein Ib/IX/V.
Philadelphia chromosome Reciprocal translocation of the platelet aggregation Platelet-to-platelet binding, part of
long arm of chromosome 22 to chromosome 9; definitive a sequential mechanism leading to the initiation and
for the diagnosis of chronic myelogenous leukemia. The formation of a thrombus or hemostatic plug. Requires
mutation results in the fusion of the BCR and ABL genes fibrinogen and platelet membrane receptor glycoprotein
and abnormal tyrosine kinase production. IIb/IIIa.
836 APPENDIX Glossary

platelet factor 4 (PF4) Protein released from platelet polyclonal Describes a group of cells or organisms derived
-granules that binds and inhibits heparin. The heparin-PF4 from several cells. Each cell is identical to its parent cells
complex is implicated in heparin-induced thrombocytope- (i.e., a clone), but since all cells of the group did not derive
nia with thrombosis. from the same cell, the group of cells is mixed. A polyclonal
platelet-poor plasma (PPP) Plasma centrifuged to achieve antibody is developed within a laboratory animal and not
a platelet count of less than 10,000/mcL. PPP is required for a hybridoma.
all coagulation testing, and only PPP may be frozen. polycythemia (erythrocytosis) Elevated red blood cell
platelet-rich plasma (PRP) Plasma centrifuged at 50 xg to count, hemoglobin, and hematocrit in peripheral blood,
achieve a platelet count of 200,000/mcL to 300/000/mcL. usually in response to chronic hypoxia.
Platelet-rich plasma is used for light-transmission platelet polycythemia vera (PV) Myeloproliferative neoplasm in
aggregometry. which a somatic mutation leads to a marked increase in the
platelet satellitosis (satellitism) Antibody-mediated in vitro red blood cell (RBC) count, hematocrit, hemoglobin, white
adhesion of platelets to segmented neutrophils. Occurs pri- blood cell count, platelet count, and total blood volume.
marily in specimens anticoagulated with ethylenediaminete RBC precursors are hypersensitive to erythropoietin.
traacetic acid (EDTA) and causes pseudothrombocytopenia. polymerase chain reaction (PCR) Laboratory process in
pleomorphic Occurring in various distinct forms; having the which a strand of DNA is replicated in a thermocycler to
ability to exist in various forms and to change from one produce millions of copies within a few hours.
form to another. polymorphonuclear neutrophil (PMN, segmented
pluripotential stem cell Stem cell that has the potential to neutrophil, seg) White blood cell whose nucleus is con-
differentiate into one of several types of progenitor cells, densed into two to five segments connected by filaments.
including lymphocytes, monocytes, granulocytes, thrombo- Distinguished from mononuclear cells such as monocytes
cytes, red blood cells, and nonhematopoietic stem cells. and lymphocytes.
pneumatic tube system System of tubes for transporting polyploid Having more than the characteristic diploid set of
blood specimens or other small materials in hospitals by chromosomes; for example, triploid (3), tetraploid (4),
forced air. and so on.
poikilocytosis Presence in the peripheral blood of red blood porphyria Hereditary anemia caused by impaired heme
cell with varying or bizarre shapes. synthesis with the accumulation of porphyrin and its pre-
point-of-care (POC) testing Rapid-turnaround clinical cursors. Acquired porphyria may be seen in acute lead
tests performed outside of the clinical laboratory at or near poisoning.
the patient; usually performed by nonlaboratory personnel porphyrin Product of the metabolism of a group of iron- or
but managed for quality by laboratory personnel. magnesium-free pyrrole derivatives such as protoporphyrin
polychromatic (polychromatophilic) Having a staining and protoporphyrinogen that incorporates ferrous iron to
quality in which both acid and basic stains are incorporated. form heme.
Usually used to denote a mixture of pink and blue in posttransfusion purpura Antibody-induced thrombocyto-
Wright-stained specimens. penia in patients who have received multiple transfusions
polychromatic normoblast (polychromatophilic nor- of red blood cells or platelet concentrate. Antibodies to
moblast, rubricyte) Precursor in the erythrocytic matura donor platelets cross-react with patient platelets to cause
tion series, intermediate between the basophilic normoblast potentially life-threatening thrombocytopenia.
(prorubricyte) and the orthochromic normoblast (metaru- postmenstrual age Time elapsed between the first day of the
bricyte). In this stage, differentiation is based on the decreas- mothers last menstrual period and birth (gestational age)
ing cell diameter and the gray-blue cytoplasm as hemoglobin plus the time elapsed after birth (chronologic age). For
first becomes visible. example, a preterm infant born at a gestational age of 32
polychromatic or polychromatophilic red blood cell weeks who is currently 10 weeks old (chronologic age)
(reticulocyte) Immature but anucleate red blood cell would have a postmenstrual age of 42 weeks.
(RBC) with grayish cytoplasm on a Wright-stained blood precision Degree to which the results of replicate analyses of
film. When new methylene blue dye is used, the cytoplasm a sample parallel each other, often expressed as coefficient
of these cells has a meshlike pattern of dark blue threads and of variation or percent coefficient of variation.
particles, vestiges of the endoplasmic reticulum. Reticulocy- precursor Differentiating (immature) bone marrow cell stage
tosis or polychromatophilia indicates bone marrow regen- that is morphologically identifiable as belonging to a given
eration activity in hemolytic anemia or chronic blood loss. cell line; for example, pronormoblasts (rubriblasts) are pre-
polychromatophilia (reticulocytosis) Elevated reticulo- cursors of basophilic normoblasts (prorubricytes) in eryth-
cyte count on a peripheral blood film stained with new ropoiesis. Precursor may also refer to the inactive zymogen
methylene blue dye or increase in the number of polychro- forms of coagulation factors; for example, prothrombin is a
matophilic red blood cells on a Wright-stained blood precursor of thrombin.
film. Reticulocytosis or polychromatophilia indicates bone preeclampsia Pathologic condition of late pregnancy charac-
marrow regeneration activity in hemolytic anemia or terized by edema, proteinuria, and hypertension; may lead
chronic blood loss. to eclampsia, which presents with seizures and is often fatal.
APPENDIX Glossary 837
prekallikrein (PK, pre-K, Fletcher factor) Member of enabling activated protein C, a serine protease, to inactivate
the kinin inflammatory system that forms active kallikrein factors Va and VIIIa.
upon digestion by kininogen. Pre-K helps to trigger in vitro proteins in vitamin K antagonism (PIVKA) Name given
coagulation contact activation. Pre-K deficiency prolongs to the forms of factors II, VII, IX, and X and proteins C, S,
the partial thromboplastin time but has no clinical and Z that, under conditions of vitamin K absence or antag-
consequence. onism, lack a second glutamic acid carboxyl group as
primary hemostasis First stage in coagulation in which the required for binding to Ca2+ and phospholipid. Also called
blood vessels contract to seal the wound and platelets des--carboxyl coagulation factors. The des--carboxyl forms of
and von Willebrand factor fill the open space by forming a these factors cannot participate in coagulation reactions.
platelet plug. Vitamin K antagonism is the basis for oral anticoagulant
primary standard Reference material that is of fixed and therapy with warfarin (Coumadin).
known composition and is capable of being prepared in proteolytic Pertaining to any substance that digests protein
essentially pure form. by hydrolyzing primary peptide bonds.
primer Short piece of synthetic DNA complementary to a prothrombin (coagulation factor II) Plasma precursor of
target DNA sequence. The primer acts as a point from which the coagulation factor thrombin. It is converted to thrombin
replication can proceed, as in a polymerase chain reaction. by activated factor X complexed to factor V.
procoagulant Coagulation (clotting) factor. During the prothrombin time (protime, PT) Test to measure the
coagulation process, inactive procoagulants become acti- activity of coagulation factors I (fibrinogen), II (prothrom-
vated to form serine proteases or cofactors and function bin), V, VII, and X, which participate in the extrinsic and
together to produce a localized thrombus. common pathways of coagulation. Thromboplastin and
progenitor Undifferentiated (immature) hematopoietic cell calcium are added to plasma and the clotting time is
that is committed to a cell line but cannot be identified recorded. Widely used to monitor warfarin therapy.
morphologically. proto-oncogene Normal gene controlling the cell cycle
prolymphocyte Developmental form in the lymphocytic and cell division that, upon activation, may become an
series that is intermediate between the lymphoblast and the oncogene.
lymphocyte. pseudoPelger-Hut cell (Pelgeroid cell) Hyposegmented,
promegakaryocyte Morphologically identifiable bone hypogranular neutrophils that resemble Pelger-Hut cells.
marrow cell stage that is intermediate between the Helpful in the diagnosis of leukemia, myeloproliferative
megakaryoblast and the megakaryocyte. neoplasms, and myelodysplastic syndromes.
promonocyte Precursor in the monocytic series; the cell pseudoleukocytosis Falsely increased white blood cell
stage intermediate in development between the monoblast (WBC) count indicating mobilization of the marginal WBC
and the monocyte. pool caused by strenuous physical activity, cold, or fever.
promyelocyte Precursor in the granulocytic (myelocytic) pseudothrombocytopenia Falsely decreased platelet count
series that is intermediate in development between a myelo- often caused by platelet satellitosis.
blast and a myelocyte; contains primary granules. pulmonary embolism (PE) Pathologic movement of a
pronormoblast (rubriblast) Undifferentiated (immature) proximal fragment of clot from a deep vein thrombosis
hematopoietic cell that is the most primitive morphologi- through the right side of the heart to the pulmonary circula-
cally identifiable precursor in the erythrocytic series; dif- tion, where it lodges in an artery and causes infarction of
ferentiates into the basophilic normoblast (prorubricyte). the lung. Approximately one third of pulmonary embolisms
prorubricyte (basophilic normoblast) Second identifi- are fatal within 1 hour.
able stage in bone marrow erythrocytic maturation; it is purpura Purple skin discoloration of 1cm or greater diam-
derived from the pronormoblast (rubriblast). Typically 10 eter seen in mucocutaneous bleeding. Usually caused by
to 12m in diameter, the prorubricyte has cytoplasm that thrombocytopenia, platelet disorders, von Willebrand
stains dark blue with Wright stain. disease, or vascular disorders such as scurvy.
prostaglandin (PG) Any of a family of unsaturated 20- pyknosis Degeneration of a cell in which the nucleus shrinks
carbon fatty acids that are cleaved from cell membrane in size and the chromatin condenses to a solid, structureless
phospholipids and serve as intracellular activators and mass or masses. Part of the process of apoptosis, or indica-
inhibitors. The prostaglandins include thromboxane A2, a tive of the effects of chemotherapy.
platelet activator, and prostacyclin, a platelet inhibitor pro- pyropoikilocytosis (hereditary pyropoikilocytosis)
duced by endothelial cells. Rare hereditary defect of spectrin that causes severe hemo-
protein C (PC) Vitamin Kdependent coagulation control lytic anemia beginning in childhood with extreme poikilo-
protein. A plasma serine protease activated by thrombin- cytosis in which the red blood cell morphology resembles
thrombomodulin and stabilized by its cofactor, protein S, that seen in burn patients.
protein C inhibits coagulation by inactivating factors Va pyruvate kinase (PK) Enzyme that converts pyruvate to
and VIIIa. lactate; essential for anaerobic glycolysis.
protein S (PS) Vitamin Kdependent coagulation control pyruvate kinase deficiency Deficiency of pyruvate kinase,
protein. Serves as a stabilizing cofactor for protein C, the enzyme that converts pyruvate to lactate, which causes
838 APPENDIX Glossary

hemolytic anemia by reducing red blood cell life span. It the standard deviation. Each laboratory must define the
is the most common enzyme deficiency of the Embden- reference interval for the instrument that is being used and
Meyerhof pathway. for the population that is being served.
Q arm Long arm of a chromosome. refractive index Speed at which light travels in air, divided
Q banding (quinacrine banding) In cytogenetic analysis, by the speed at which light travels through another medium,
a procedure in which metaphase chromosomes are such as immersion oil. The refractive index of oil is similar
stained with fluorescent quinacrine dye. The areas rich in to the refractive index of air.
adenine-thymine, called Q+, fluoresce intensely, whereas reliability Extent to which a method is able to maintain both
the areas rich in guanine-cytosine (Q) fluoresce more accuracy and precision over a defined period of time.
lightly. The Q+ bands correspond with G bands in Giemsa remission Partial or complete disappearance of the clinical
stainbased G banding. Banding patterns are used to iden- and laboratory characteristics of a chronic or malignant
tify chromosomes. disease.
qualitative analysis Study of a sample to determine the replication DNA duplication or synthesis prior to mitosis;
presence, but not the concentration, of specific chemicals. may also be used to refer to mitosis.
quality control Term used to refer to the control and moni- reptilase Thrombinlike enzyme isolated from the venom of
toring of the testing process to ensure that the results are Bothrops atrox that catalyzes the conversion of fibrinogen to
valid and reproducible. fibrin in a manner similar to thrombin.
quantitative analysis Determination of the concentration resheathing device Device that allows safe one-handed
of an analyte. recapping of a hypodermic blood collection needle to
radiotherapy Treatment of neoplastic disease using x-rays or prevent needle stick injury.
gamma rays to deter the proliferation of malignant cells by resolution In microscopy, the smallest distance between
disrupting mitosis or impairing DNA synthesis. which two adjacent objects can be distinguished. A measure
Raynaud phenomenon (acrocyanosis) Persistent sym- of image and lens quality that relates to the smallest feature
metrical cyanosis (blotchy blue or red discoloration) of the that can be seen with a set of lenses.
skin of the digits, palm, wrists, and ankles upon prolonged restriction endonuclease Enzyme that cleaves DNA at a
exposure to cold. specific nucleotide site.
reactive lymphocytes (atypical, transformed, or variant reticulocyte (polychromatic or polychromatophilic
lymphocytes) Lymphocytes whose altered morphology red blood cell) Immature but anucleate red blood cell
includes stormy blue cytoplasm and lobular or irregular (RBC) that shows a meshlike pattern of dark blue threads
nuclei. Variant lymphocytes indicate stimulation by a virus, and particles, vestiges of the endoplasmic reticulum, when
particularly Epstein-Barr virus, which causes infectious stained with new methylene blue vital dye. In a Wright-
mononucleosis. stained blood film, no filaments are seen but the cytoplasm
recessive Denoting an allele whose product or effect is stains grayish and the cell is called a polychromatic or poly-
masked by a dominant allele at the corresponding locus. chromatophilic RBC. Reticulocytosis or polychromatophilia
When an individual has two recessive genes, he or she is indicates bone marrow regeneration activity in hemolytic
homozygous recessive, and the trait is expressed. anemia or chronic blood loss.
red blood cell (RBC) indices Numerical representations reticulocyte production index (RPI) Index calculated to
of average RBC volume (mean corpuscular volume), hemo- correct for the presence of shift reticulocytes that otherwise
globin mass (mean corpuscular hemoglobin), relative may falsely elevate the visual reticulocyte count.
hemoglobin concentration (mean corpuscular hemoglobin reticulocytosis (polychromatophilia) Elevated reticulo-
concentration), and variation in RBC volume, called the red cyte count on a peripheral blood film stained with new
cell distribution width. Indices are computed from the RBC methylene blue dye or increase in the number of polychro-
count, and hemoglobin and hematocrit values are directly matophilic red blood cells on a Wright-stained blood
measured by hematology analyzers. film. Reticulocytosis or polychromatophilia indicates bone
red cell distribution width (RDW) Coefficient of varia- marrow regeneration activity in hemolytic anemia or
tion of red blood cell volume. An increased red cell distribu- chronic blood loss.
tion width indicates anisocytosis. reticuloendothelial system (RES) System of fixed and
red marrow Hematopoietic bone marrow, in contrast to motile phagocytic macrophages and their precursor mono-
yellow, fatty bone marrow. cytes engaged in primary efferent immunity. Macrophages
Reed-Sternberg cell Giant, typically binucleate cell whose are found in every organ and tissue.
halves are mirror images. The nuclei are enclosed in abun- retrovirus Any of a family of RNA viruses containing the
dant cytoplasm and contain prominent nucleoli. The pres- enzyme reverse transcriptase.
ence of Reed-Sternberg cells is the definitive histiologic reverse transcription polymerase chain reaction (RT-
characteristic of Hodgkin disease. PCR) Polymerase chain reaction amplification process
reference interval (RI) Range of test results for a given that produces complementary DNA from the messenger
analyte that is seen in a healthy population of individuals, RNA present in an RNA sample extracted from patient
typically computed as the mean plus or minus two times cells.
APPENDIX Glossary 839
Rh immune globulin (RhIg) Solution containing anti of points provides an indication of the relationship between
bodies specific for the Rh(D) antigen given intramuscularly the two variables. In hematology a typical scatterplot graphs
to an Rh(D)-negative mother with an Rh(D)-positive fetus cell size against cytoplasmic complexity.
or infant to prevent sensitization of the mother to the schistocyte (schizocyte) Fragmented red blood cell charac-
infants D antigen. It is given postpartum within 72 hours teristic of microangiopathic hemolytic anemia, severe burns,
of delivery of a D-positive infant or antepartum at 28 weeks disseminated intravascular coagulation, and prosthetic
gestation and again at delivery. mechanical trauma.
Rh-null disease Hemolytic anemia in persons who lack all scurvy Deficiency of vitamin C (ascorbic acid) causing con-
Rh antigens (Rh null); marked by spherocytosis, stomato- nective tissue breakdown. Marked by weakness, anemia,
cytosis, and increased osmotic fragility. spongy gums, and a tendency to mucocutaneous bleeding.
ribonucleic acid (RNA) Single strand of polynucleotides secondary hemostasis Second phase of coagulation involv-
connected by ribose molecules. RNA base sequences are ing the activation of plasma coagulation proteins to produce
transcribed from DNA and are the basis for translation to a fibrin clot.
proteins. Types of RNA include heteronuclear RNA, mes- secondary standard (calibrator) Calibration material for
senger RNA, ribosomal RNA, and transfer RNA. which the analyte concentration has been ascertained by
ribosomes Granules embedded in the membranes of endo- reference to a primary standard or by controlled reference
plasmic reticulum that are composed of protein and RNA. assays.
Ribosomes are the sites for primary protein translation from segmented neutrophil (seg, polymorphonuclear neu-
messenger and transfer RNA. trophil, PMN) White blood cell whose nucleus is con-
ringed sideroblast Nucleated red blood cell precursor with densed into two to five segments connected by filaments.
six or more iron granules that circle at least one third of the Distinguished from mononuclear cells such as monocytes
nucleus. These cells, visible with Prussian blue stain, are the and lymphocytes.
pathognomonic finding in refractory anemia with ringed selectin Any of a family of cell adhesion molecules that
sideroblasts. mediate the binding of white blood cells and platelets to
ristocetin Antibiotic no longer in clinical use that facilitates the vascular endothelium.
the in vitro interaction of von Willebrand factor with plate- senescent Aging or growing old. A senescent red blood cell
let membrane glycoprotein Ib/IX/V. Used as an agonist in loses its deformability and is cleared by the spleen.
platelet aggregation to test for von Willebrand disease. sensitivity In laboratory testing, the conditional probability
Romanowsky stain Prototype of the many eosinmethylene that a person with a given disease will be correctly identi-
blue stains for blood cells and malarial parasites, including fied as having it by a clinical test (i.e., diagnostic sen
Wright and Giemsa stain. sitivity). Also, the lowest level of a substance that can be
rouleaux Aggregation of stacked red blood cells caused detected by a laboratory test procedure (i.e., analytical
by elevated plasma proteins and abnormal monoclonal sensitivity).
proteins. sepsis (septicemia) Proliferation of pathologic organisms
rubriblast (pronormoblast) Undifferentiated (immature) in the blood.
hematopoietic cell that is the most primitive morphologi- sequestration Transfer of blood cells from the circulation
cally identifiable precursor in the erythrocytic series; dif- into a limited vascular area, such as the spleen. Platelets may
ferentiates into the basophilic normoblast (prorubricyte). be sequestered, which results in a decrease in their circulat-
rubricyte (polychromatic or polychromatophilic nor- ing numbers.
moblast) Precursor in the erythrocytic maturation series serine protease Any of a group of proteolytic enzymes of the
that is intermediate between the basophilic normoblast trypsin family that include activated procoagulants (throm-
(prorubricyte) and the orthochromic normoblast (metaru- bin and factors VIIa, IXa, Xa, XIa, and XIIa) and activated
bricyte). In this stage, differentiation is based on the decreas- inhibitors (antithrombin, activated protein C). Serine pro-
ing cell diameter and the gray-blue cytoplasm as hemoglobin teases are synthesized as inactive zymogens; activation
first becomes visible. occurs when the zymogen is cleaved at one or more specific
Russell bodies Plasma cell cytoplasmic inclusions that sites by the action of another protease during the coagula-
appear similar to vacuoles containing aggregates of tion process.
immunoglobulins. Plasma cells with Russell bodies are serine protease inhibitor (serpin) Plasma proteins, for
called Mott cells. instance antithrombin and heparin cofactor II, which
Russell viper venom time Rarely use coagulation assay control the coagulation cascade by inhibiting the serine
similar to the prothrombin time. Russell viper venom acti- proteases, particularly factors IIa and Xa.
vates the coagulation factor common pathway at the level serotonin Potent vasoconstrictor released from the -granules
of factor X. The dilute Russell viper venom time assay is used of activated platelets.
routinely in lupus anticoagulant screening. Szary cell Mononuclear cell with a cerebriform nucleus
scatter plot Plot on rectangular coordinates of paired obser- (resembling the surface of the cerebrum) and a narrow rim
vations of two random variables, with each observation of cytoplasm. It is a characteristic finding in cutaneous T-cell
plotted as one point on the graph; the scatter or clustering lymphomas.
840 APPENDIX Glossary

Szary syndrome Cutaneous T-cell lymphoma characterized have a reduced diameter. Spherocytes appear most fre-
by exfoliative erythroderma, peripheral lymphadenopathy, quently in warm autoimmune hemolytic anemia and
and the presence of Szary cells in the skin, lymph nodes, hereditary spherocytosis.
and peripheral blood. spleen Large organ in the upper left quadrant of the abdomen,
shift reticulocyte Gray-blue red blood cell with increased just under the stomach. The spleen has the bodys largest
diameter. The shift reticulocyte is a reticulocyte that has collection of macrophages, which are responsible for phago-
been released from the bone marrow prematurely to com- cytosis and elimination of senescent red blood cells. The
pensate for hemolytic anemia or chronic blood loss. Shift spleen also houses multiple lymph nodes.
reticulocytes require more than 1 day in the peripheral splenectomy Excision of the spleen.
blood to lose residual RNA and gain a mature-looking splenomegaly Enlargement of the spleen.
reddish cytoplasm. standard deviation Mathematic expression of the disper-
sickle cell (drepanocyte) Abnormal crescent-shaped red sion of a set of values or scores about the mean.
blood cell containing hemoglobin S, characteristic of sickle standard precautions (universal precautions) Practices
cell anemia. to control bloodborne disease in which all human blood
sickle cell anemia (sickle cell disease) Severe chronic and body fluids are treated as if infectious. Infection is
hemoglobinopathy in people who are homozygous for prevented by a series of protective methods including the
hemoglobin S. The abnormal hemoglobin results in distor- use of sterile gloves, fluid-impermeable clothing, and eye
tion of red blood cells (sickle cells) and leads to crises protection.
characterized by joint pain, anemia, thrombosis, fever, and steatorrhea Fat in the stool, usually due to malabsorption.
splenomegaly. Stool is green or colorless and foul smelling.
sickle cell crisis Any of several acute conditions occurring as stem cell Undifferentiated mononuclear bone marrow or
part of sickle cell disease, such as aplastic crisis, which is peripheral blood cell whose daughter cells may give rise to
temporary bone marrow aplasia; hemolytic crisis, which is a variety of cell types and which is capable of renewing itself
acute red blood cell destruction; and vasoocclusive crisis, and thus maintaining a pool of cells that can differentiate
which is severe pain due to blockage of the blood vessels. into multiple other cell types.
sickle cell trait Asymptomatic heterozygous form of sickle stomatocyte Abnormal cup-shaped mature red blood cell
cell anemia characterized by the presence of both hemoglo- that has a slitlike area of central pallor.
bin S and hemoglobin A. storage pool deficiency Inadequacy of platelet -granules
sideroblast Bone marrow erythrocytic precursor that shows or -granule contents that causes mucocutaneous bleeding.
excessive iron granules (siderotic granules) with Prussian Usually hereditary and related to conditions with oculocu-
blue staining. taneous albinism, such as Hermansky-Pudlak syndrome,
siderocyte Nonnucleated red blood cell in which particles of Chdiak-Higashi syndrome, and Wiskott-Aldrich syndrome.
iron (siderotic granules) are visible with Prussian blue Acquired storage pool disorder is sometimes associated with
staining. myelodysplastic syndrome.
siderotic granules (Pappenheimer bodies) Red blood stroma Supporting tissue or matrix of an organ.
cell inclusions composed of ferric iron. With Prussian blue subcutaneous (SC) Injected within the subdermal or
iron staining, they appear as multiple dark blue irregular dermal layer.
granules at the periphery. With Wright staining they appear sulfhemoglobin Hemoglobin with a sulfur atom on one of
as pale blue clusters. its porphyrin rings, which makes it ineffective for transport-
Southern blotting Technique in which DNA fragments sep- ing oxygen. Results from ingestion of sulfa-type antibiotics.
arated by gel electrophoresis are transferred to a nitrocel- supernatant Clear upper liquid part of a suspension after it
lulose filter on which specific fragments can then be detected has been centrifuged.
by their hybridization to probes. suppuration Formation or discharge of pus.
specificity In laboratory testing, the conditional probability supravital stain (vital stain) Stain that colors living tissues
that a person who does not have a specific disease will be or cells.
correctly identified as not having it by a clinical test (i.e., surface-connected canalicular system (SCCS, open
diagnostic specificity). Also used to describe the attribute of canalicular system, OCS) System of channels that is
antibodies that are able to bind only with the antigen that distributed throughout platelets and extends the plasma
stimulated their production. membrane inward. The SCCS binds numerous coagulation
spectrin Fibrous protein attached to glycophorin at the cyto- factors and provides a route for secretion of the protein
plasmic surface of the cell membrane, providing cytoskeletal contents of -granules.
support to the membrane and thus maintaining its shape. syncope Brief lapse in consciousness; fainting.
Abnormalities in red blood cell spectrin account for heredi- systemic bleeding disorder Chronic episodic bleeding
tary spherocytosis, ovalocytosis, and pyropoikilocytosis. that is evidenced by bilateral petechiae and purpura, epi-
spherocyte Abnormal spherical red blood cell with a high staxis, hematemesis, and menorrhagia. Indicates a primary
cytoplasm-to-membrane ratio. In Wright-stained peripheral coagulopathy such as thrombocytopenia, von Willebrand
blood films, spherocytes are dense, lack central pallor, and disease, or a qualitative platelet disorder.
APPENDIX Glossary 841
systemic lupus erythematosus (SLE) Chronic autoim- thrombotic thrombocytopenic purpura (TTP) Congen-
mune inflammatory disease manifested by severe vasculitis, ital or acquired deficiency of ADAMTS 13, an endothelial
renal involvement, and lesions of the skin and nervous cell von Willebrand factorcleaving protease. Ultra-large
system. von Willebrand factor multimers activate platelets to form
T cell (T lymphocyte) Lymphocyte that participates in cel- white clots in the microvasculature, causing severe throm-
lular immunity, including cell-to-cell communication. The bocytopenia with mucocutaneous bleeding, microangio-
major T-cell categories are helper cells and suppressor- pathic hemolytic anemia, and neuropathy.
cytotoxic cells. thromboxane A2 Metabolically active product of the eico-
target cell (codocyte) Poorly hemoglobinized red blood sanoid synthesis (cyclooxygenase, prostaglandin) pathway
cell (RBC) that is present in hemoglobinopathies, tha in platelets; binds platelet membrane receptor and activates
lassemia, and liver disease. In a Wright-stained peripheral platelets.
blood film, hemoglobin concentrates in the center thromboxane B2 Metabolically inactive measurable plasma
of the RBC and around the periphery to resemble a product of the eicosanoid synthesis (cyclooxygenase, pros-
bulls-eye. taglandin) pathway.
teardrop cell (dacryocyte) Red blood cell with a single thrombus In vivo blood clot causing vascular occlusion and
pointed extension, resembling a teardrop. Dacryocytes are tissue ischemia.
often seen in the myeloproliferative neoplasm called myelo- tissue factor (TF) Constitutive membrane protein of the
fibrosis with myeloid metaplasia. subendothelium. Exposure to tissue factor activates factor
telangiectasis Permanent dilation of capillaries, arterioles, VII and the tissue factor (extrinsic) coagulation pathway.
and venules that creates focal red lesions, usually in the skin Tissue factor is expressed on monocytes and endothelial
or mucous membranes. cells in chronic inflammation.
telomere Polyadenine chromosome terminus. tissue plasminogen activator (TPA) Serine protease that
tetraploid Possessing a double diploid chromosome comple- is secreted by endothelial cells and binds fibrin. It activates
ment, or four of each chromosome (4N). nearby plasminogen molecules to trigger fibrinolysis, with
thalassemia Production and hemolytic anemia characterized the formation of fibrin degradation products.
by microcytic, hypochromic red blood cells caused by defi- toxic granulation Presence of abnormally large, dark-
cient synthesis of - or -globin chains. staining, or dominant primary granules in neutrophils
thrombin Primary serine protease of coagulation. Factors Xa associated with bacterial infections.
and Va combine to cleave prothrombin to produce throm- transcription Process by which messenger RNA is produced
bin. Thrombin cleaves fibrinopeptides A and B from fibrino- from a DNA template.
gen to initiate fibrin polymerization. Thrombin potentiates transferrin Plasma iron-transport protein that moves iron
coagulation by activating platelets and factors XI, VIII, V, from sites of absorption and storage to hematopoietic tissue
and XIII and also activates the coagulation control protein, for incorporation into rubriblasts.
protein C. transformed lymphocytes (atypical, variant, or reactive
thrombin clotting time (TCT, thrombin time, TT) Coag- lymphocytes) Lymphocytes whose altered morphology
ulation test that measures the interval to clot formation after includes stormy blue cytoplasm and lobular or irregular
the addition of thrombin to plasma. Often used to test for nuclei. Variant lymphocytes indicate stimulation by a virus,
the presence of heparin. particularly Epstein-Barr virus, which causes infectious
thrombocyte Platelet. mononucleosis.
thrombocythemia Abnomally high platelet count with dys- translation Process by which the genetic information carried
functional platelets; seen in the myeloproliferative neo- by nucleotides in messenger RNA directs the sequence of
plasm known as essential thrombocythemia. amino acids in the synthesis of a specific polypeptide.
thrombocytopenia Platelet count below the lower limit of translocation Rearrangement of DNA within a chromosome
the reference interval, usually 150,000/mcL. or transfer of a segment of one chromosome to a nonho-
thrombocytosis Platelet count above the upper limit of the mologous one.
reference interval, usually 450,000/mcL. triploid Possessing a single addition chromosome comple-
thrombophilia (hypercoagulability) Abnormally increased ment, resulting in three of each chromosome (3N).
tendency to clotting caused by a number of acquired or con- trisomy Presence of an extra chromosome in addition to
genital factors. a homologous pair; for example, trisomy 21 in Down
thrombopoietin Hormone produced by renal tissue that syndrome.
recruits stem cells to the megakaryocyte cell maturation line tungsten-halogen light bulb Bulb used in brightfield
and stimulates megakaryocyte mitosis and maturation in microscopy consisting of a tungsten filament enclosed in a
response to thrombocytopenia. small quartz bulb filled with halogen gas. Tungsten creates
thrombosis Formation, development, or presence of a clot a very bright yellow light.
in a blood vessel (i.e., a thrombus). unfractionated heparin (UFH, standard heparin)
thrombospondin Adhesive glycoprotein secreted by endo- Heparin extracted from porcine mucosa and purified to
thelial cells and platelet -granules. yield molecular weights ranging from 7000 to 15,000. Used
842 APPENDIX Glossary

routinely in cardiac surgery. Unfractionated heparin therapy waived testing Test classification defined by the Clinical
requires monitoring with the partial thromboplastin time Laboratory Improvement Amendments which includes tests
or activated clotting time assay. that are simple and accurate and can be performed by non-
universal precautions (standard precautions) Practices certified personnel.
to control blood-borne disease in which all human blood Waldenstrm macroglobulinemia Form of monoclonal
and body fluids are treated as if infectious. Infection is gammopathy in which IgM is overproduced by the clone of
prevented by a series of protective methods including the a plasma cell. Increased viscosity of the blood may result
use of sterile gloves, fluid-impermeable clothing, and eye in circulatory impairment, and normal immunoglobulin
protection. synthesis is decreased, which increases susceptibility to
urobilinogen Colorless water-soluble compound formed in infections.
the intestine through the breakdown of bilirubin by bacte- warfarin (Coumadin, oral anticoagulant therapy, OAT)
ria; may appear in the urine. Vitamin K antagonist used as an anticoagulant to prevent
urokinase Enzyme produced by the kidney endothelial cells thrombosis in people with atrial fibrillation, venous
that acts as a plasminogen activator. thromboembolism, or cardiac insufficiency. Also used
vacuole Any clear space or cavity formed in the cytoplasm of prophylactically after orthopedic surgery. Warfarin sup-
a cell. presses vitamin K and reduces the activity of the vitamin
vacuolization Formation of vacuoles. Kdependent coagulation factors II (prothrombin), VII, IX,
vasoconstriction Reduction in blood vessel diameter due to and X.
smooth muscle constriction. warm antibody IgG antibody that reacts optimally at a tem-
venipuncture Use of a hypodermic needle to puncture a vein perature of 37 C.
and collect blood. warm autoimmune hemolytic anemia Most common
vertigo Sensation of rotation or movement of oneself or ones autoimmune hemolytic anemia, which results from the
surroundings; dizziness. reaction of IgG autoantibodies with red blood cells at an
viscosity Resistance of a liquid to flow. optimal temperature of 37 C.
vital stain (supravital stain) Stain that colors living tissues Wiskott-Aldrich syndrome Immunodeficiency disorder
or cells. characterized by oculocutaneous albinism, thrombocytope-
vitamin B12 (cyanocobalamin) Complex vitamin involved nia, inadequate T- and B-cell function, and an increased
in the metabolism of protein, fats, and carbohydrate; susceptibility to viral, bacterial, and fungal infections.
normal blood formation; and nerve function. xanthochromic Having a yellowish color. Used to describe
vitamin K Natural phylloquinone occurring in green leafy cerebrospinal fluid, in which xanthochromia indicates the
vegetables and liver and produced by commensal intestinal presence of bilirubin and thus serves as evidence of a prior
organisms. Vitamin K catalyzes the -carboxylation of glu- episode of bleeding into the brain.
tamic acid in a number of calcium-binding proteins, includ- X-linked Pertaining to genes or to the characteristics
ing the vitamin Kdependent coagulation proteins factors or conditions they transmit that are carried on the X
II (prothrombin), VII, IX, and X, and control proteins C, S, chromosome.
and Z. X-linked recessive inheritance Pattern of inheritance in
vitamin K antagonist (VKA) Substance that inhibits the which a recessive allele is carried on the X chromosome;
action of vitamin K; for example, warfarin (Coumadin), results in the carrier state in females and development of
which is used in oral anticoagulant therapy. disease characteristics in males, since they do not have a
von Willebrand disease Congenital autosomal dominant normal X chromosome to compensate.
variable mucocutaneous bleeding disorder characterized by zymogen Inactive precursor that is converted to an active
a deficiency of von Willebrand factor activity and antigen, form by an enzyme. Zymogens are inactive coagulation
and subsequent impairment of platelet adhesion. factors, such as prothrombin.
Index
Acute leukemia, 3, 291t, 546-557 ADP. See Adenosine diphosphate (ADP).
A of ambiguous lineage, 555 Aerobic glycolysis, 105f, 106, 106t
Abbott hematology analyzers, 602t, 606-608, case study on, 546, 546f Afibrinogenemia, 663t, 727-728
609f-610f chromosomal abnormalities in, 455b, 456-457, AG-NOR-banding stain, 449
Abciximab, 159-160, 776-777 456f-457f Agammaglobulinemia, X-linked, 415
platelet dysfunction from, 725 Acute lymphoblastic leukemia, 547-549 Agarose gel, 476
Abetalipoproteinemia, 324 chromosomal abnormalities in, 456, 456f Age. See also Children; Elderly.
ABL protein, 510-511 cytochemical stain reaction chart for, 440t gestational, hematologic values and, 581
ABL1-BCR fusion gene. See BCR-ABL1 fusion gene. flow cytometry in, 547-548 thrombosis risk and, 670t
ABO blood group genetics of, 548 Aggregation, platelet, 163-164, 164f-165f, 629-630,
accidental transfusion of, hemolytic reaction to, morphology of, 547, 547f 630f
361 Philadelphia chromosomepositive, 512-514, 548 Aggregometer, 741-742, 742f
von Willebrand factor reference intervals for, 658, prognosis in, 548 Aggregometry, 741-747
658t treatment of, 548-549 in heparin-induced thrombocytopenia, 688, 702,
ABO hemolytic disease of fetus and newborn, 362, WHO classification of, 547, 547b 747
362t Acute megakaryoblastic leukemia in liver disease, 651t
Acanthocytes, 245t, 247f flow cytometry in, 497 low-dose ristocetin-induced, 657, 657t, 745-746
membrane defects with, 324-325 with t(1;22)(p13;q13)(RBM15-MLK1), 551 lumi-, 165-166, 688, 742, 744f, 745
Acanthocytosis Acute megakaryocytic leukemia, 554, 554f platelet agonists for, 742-747, 743t, 746t
chorea, 324-325 Acute monoblastic leukemia, 553, 553f platelet-rich plasma for, 740, 742, 742f-743f
hemolytic anemia with, 324 Acute monocytic leukemia, 440t, 553, 553f in storage pool disorders, 746, 746t
neurologic impairment and, 324-325 Acute myeloid leukemia, 549-555 in von Willebrand disease, 657, 657t, 745-746
Accuracy, 42-44, 43f-44f chromosomal abnormalities in, 457, 457f whole-blood specimens for, 740, 742, 744f
Acetic acid, for body fluid cell counts, 227, 228t classification of, 455, 549-555, 549b Agkistrodon contortrix venom activator, 679, 679f
Acetylsalicylic acid. See Aspirin (acetylsalicylic acid). clinical presentation in, 549 Agranulation, in myelodysplastic syndromes, 535-536,
Achlorhydria, in pernicious anemia, 275, 278 cytochemical stain reaction chart for, 440t 536f
Achromatic lens, 33-35 with inv(3)(q21;q26.2) or t(3;3)(q21;q26.2)(RPN1- AIDS (acquired immunodeficiency syndrome)
Acid-fast stains, 220t EVI1), 551 flow cytometry in, 505
Acid phosphatase stain, 440-442, 442f, 442t with inv(16)(p13;q22) or t(16;16)(p13;q22)(CBFB- neonatal, thrombocytopenia in, 696-697
Acid sphingomyelinase deficiency, 415, 415f MYH11), 550, 550f Albinism, storage pool disorder in, 161-162
ACL coagulometers, 793t with maturation, 552, 553f Alcohol
Acquired immunodeficiency syndrome (AIDS) with minimal differentiation, 552, 552f lens cleansing with, 37
flow cytometry in, 505 with myelodysplasia-related changes, 551 skin cleansing with, 23, 27
neonatal, thrombocytopenia in, 696-697 not otherwise specified, 496-497, 498f-499f, Alcohol consumption
Acridinium ester, 480, 481f 552-554, 552t platelet function defects from, 724-725
Acrocyanosis, in cold agglutinin disease, 358 Philadelphia chromosomepositive, 513-514 thrombocytopenia and, 697
ACT. See Activated clotting time (ACT). with recurrent genetic abnormalities, 455, 455b, Alcoholic cirrhosis, platelet disorders in, 727
ACT Plus, 792t 496, 497f-498f Alder-Reilly anomaly, 410, 410f
Actin, 109f, 110, 111f, 111t with t(1;22)(p13;q13)(RBM15-MLK1), 551 Aldolase, deficiency of, 333
in platelets, 161 with t(6;9)(p23;q34)(DEK/NUP214), 551 ALK (anaplastic lymphoma kinase) protein, in
Activated clotting time (ACT), 747 with t(8;21)(q22;q22)(RUNX1/RUNX1T1), 549-550, anaplastic large cell lymphoma, 574-575
for direct thrombin inhibitor therapy monitoring, 550f Alkali denaturation test, 403
776 with t(9;11)(p22;q23)(MLLT3-MLL), 551, 551f Alkaline phosphatase, in gel electrophoresis, 478
for unfractionated heparin monitoring, 772 with t(15;17)(q22;q21)(PML-RARA), 550-551, Alkaloids, plant, for leukocyte neoplasms, 430,
Activated partial thromboplastin time (APTT), 5, 550f 431t
750-752 therapy-related, 551-552 Alkylating agents
in coagulopathy screening, 649t without maturation, 552, 552f for leukocyte neoplasms, 430, 431t
as diagnostic assay, 752 Acute myelomonocytic leukemia, 552-553, 553f myeloid neoplasms from, 551-552
for direct thrombin inhibitor therapy monitoring, cytochemical stain reaction chart for, 440t ALL. See Acute lymphoblastic leukemia.
776 with eosinophilia, 549-550, 550f Allergic disorders
in disseminated intravascular coagulation, 686, flow cytometry in, 496, 499f eosinophils in, 142
686t, 752 Acute promyelocytic leukemia, 550-551, 550f mast cells in, 144
in hemophilia, acquired, 654 chromosomal abnormalities in, 457, 457f Allergic purpura, 729
in hemophilia A, 660, 660t flow cytometry in, 496, 498f Allergy
in hemophilia B, 662 ADAMTS13, in thrombotic thrombocytopenic latex, 10
in hemophilia C, 662 purpura, 340, 707-708, 708f specimen collection and, 26
for heparin therapy monitoring, 752, 770-772, Adducin, 111t Allogeneic stem cell transplantation, 433, 433f
771f Adenine, 465, 467f-468f Alloimmune hemolytic anemia, 361-362, 362t
in liver disease, 651, 651t Adenitis, 73-74 Alloimmune neonatal thrombocytopenia, 704-705
lupus anticoagulantsensitive, 673-674, 674f, 752 Adenosine diphosphate (ADP) AMAX Destiny coagulometers, 793t
mixing studies with, 752-753 in platelet aggregometry, 743t, 745, 746t E-Aminocaproic acid, for von Willebrand disease, 658,
principle of, 750, 751f in platelets, 162t 659t
procedure for, 750-751 seven-transmembrane receptors for, 160, 160t Aminolevulinate synthase, 118, 119f, 121
quality control for, 751 Adenosine triphosphate (ATP) Aminolevulinic acid, 118, 119f, 121
reference intervals for, 751 BCR-ABL1 binding site mutations and, 515-516, Aminolevulinic acid dehydratase, 118, 119f, 121
in single-factor deficiencies, 660t, 752, 755-756, 517f AML. See Acute myeloid leukemia.
756f, 757t cation pumps dependent on, 111 Amyloidosis, 730
specimen management for, 739-741, 740t glycolysis-generated, 104, 105f, 106 Anaerobic glycolysis, 104-106, 104t, 105f
in von Willebrand disease, 654-655 in platelets, 162t Anagrelide, for essential thrombocythemia, 712-713
Activated protein C resistance, 675-676 Adenosylcobalamin, 269-270 Analytical specificity, 45-46, 51t
assay for, 676, 676f Adhesion, platelet, 159-160, 159t, 162, 163f-164f Anaplastic large cell lymphoma, 574-575, 575f,
thrombosis risk and, 671t-672t, 672, 675-676, 676f Adhesion disorders, leukocyte, 412 575t
Acute chest syndrome, in sickle cell anemia, 374 Adhesion molecules, 159-160, 159t, 629 Anaplastic lymphoma kinase (ALK) protein, in
Acute erythroid leukemia, 497, 553-554, 554f Adipocytes, 70, 211 anaplastic large cell lymphoma, 574-575
Acute erythroleukemia, 272, 553 in bone marrow specimens, 218 Androgen therapy, for dyskeratosis congenita, 290

Note: Page numbers followed by b, f, and t indicate boxes, figures, and tables, respectively.

843
844 Index

Anemia, 1, 241-267 Antibody(ies) (Continued) Area under the curve, 51, 52f
from acute blood loss and hemolysis, 243 drug-dependent, 360 Argatroban, 775, 775f
aplastic. See Aplastic anemia. drug-independent, 360 for heparin-induced thrombocytopenia, 688, 704
approach to evaluation of, 246-250 in drug-induced immune hemolytic anemia, 360 monitoring of, 776
bone marrow examination in, 244-246 hapten-dependent, in thrombocytopenia, 701 prothrombin time and, 769
case study on, 241, 253-254 in immune hemolytic anemia, 354-355, 356t Arm shield, 11f
of chronic inflammation, 259-260 monoclonal Array comparative genomic hybridization, 458, 458f
in elderly, 590 for acute lymphoblastic leukemia, 548-549 Artemether-lumefantrine, for malaria, 346-347
etiology of, 259 discovery of, 491-492 Arterial thrombosis
laboratory diagnosis of, 259-260 in flow cytometry, 492-493 management of, 766
treatment of, 260 for leukocyte neoplasms, 432 mechanism for, 669
of chronic kidney disease, 294 Anticardiolipin antibody immunoassay, 674-675 predictors of, 680-683, 680t
classification of Anticoagulant properties of vascular intima, 628-629, prevalence of, 669
mean cell volume and, 248-249, 248f-249f, 250t 628b risk of
pathophysiologic, 250, 250b Anticoagulants. See also specific agents, e.g., Warfarin. fibrinogen activity and, 680t, 682-683, 682t, 683f
red blood cell distribution width and, 249, 250t for bone marrow specimens, 215 high-sensitivity C-reactive protein and, 680-681,
reticulocyte count and, 248, 248f case study on, 765 680t-681t
clinical findings in, 242 in collection tubes, 20 homocysteine and, 672t, 680t, 681-682, 682f,
complete blood count in, 243, 244t for hemostasis specimens, 738-739, 739f 682t
Cooleys. See -Thalassemia major. in heparin-induced thrombocytopenia, 704 lipoprotein (a) and, 680t, 683, 683t
definition of, 242 lupus. See Lupus anticoagulants. Ascites, 232
Diamond-Blackfan, 292-293, 292t monitoring of, 765-782 Ascorbic acid deficiency, vascular purpura from, 730
from DNA metabolism defects, 268-282 oral, 766-767. See also Warfarin. Asparaginase, 431t
dyserythropoietic, 243 quality control for, 29 Aspartate aminotransferase, in HELLP syndrome, 341
congenital, 272, 292t, 293, 293f types of, 766 Aspiration, bone marrow. See Bone marrow aspiration.
in elderly, 590-591 Antiepileptic drugs, folate metabolism and, 273 Aspirin (acetylsalicylic acid)
in elderly, 589-591, 590t Antifibrinolytic therapy, for Glanzman antiplatelet activity of, 777, 777f
Fanconi, 288-289, 292t, 696 thrombasthenia, 721 in platelet activation, 630, 631f
general concepts in, 254 Antigen-presenting cells, 560-561 platelet function defects from, 724
in hemoglobin SC disease, 381 Antiglycolytic agents, in collection tubes, 20 for prophylaxis, 778
hemolytic. See Hemolytic anemia. Anti2-glycoprotein I immunoassay, 675 resistance to, 778
history in, 242 Antihistamines, platelet dysfunction from, 726 Aspirin-like disorder, 168, 723
in infants and children, 584 Antimalarial drugs, 346-347 AspirinWorks, 778
iron deficiency. See Iron deficiency anemia. Antiphosphatidylserine immunoassay, 675 Atherosclerosis, C-reactive protein and, 680
laboratory diagnosis of, 243-246 Antiphospholipid antibodies Atherosclerotic plaque, 669
macrocytic, 248-249, 248f-249f, 250t assays for, 673, 674f-675f Atovaquone-azithromycin, for babesiosis, 347-348
algorithm for, 279f drug-induced, 672, 673b Atovaquone-proguanil, for malaria, 346-347
classification of, 269b thrombosis risk and, 672-675 ATP. See Adenosine triphosphate (ATP).
megaloblastic. See Megaloblastic anemia. Antiphospholipid syndrome, thrombosis risk in, 670, ATR-16 syndrome, 400
nonmegaloblastic, 279 671t ATR-X syndrome, 400
marching, 257 2-Antiplasmin, 643 Autoantibodies
mean cell volume in, 243, 244t, 248-249, deficiency of, 663 in drug-induced thrombocytopenia, 701
248f-249f, 250t -Antiplasmin, 166t induction of, in erythrocyte, 360
mechanisms of, 243 Antiplatelet factor 4 (heparin-induced) antibody Autohemolysis test, in hereditary spherocytosis, 319,
megaloblastic. See Megaloblastic anemia. immunoassay, 688, 747 319f
microcytic, 248, 248f-249f, 250t Antiplatelet therapy, 776-778. See also specific agents, Autoimmune hemolytic anemia, 356-359
myelophthisic, 294 e.g., Clopidrogrel. cold, 357-359, 357t, 358f
nonmegaloblastic, 249, 249f monitoring of mixed-type, 357t, 359
normocytic, 248f-249f, 249, 250t platelet activity assays for, 778 paroxysmal cold hemoglobinuria as, 357t, 359
peripheral blood smear in, 244, 245t-246t, 247f platelet aggregometry for, 746 warm, 356-357, 357t
pernicious, 275, 278-279 platelet dysfunction from, 725-726 Automated cell-counting instrumentation, 598-625.
physical examination in, 242 resistance to, 778 See also Hematology analyzers.
physiologic, of neonate, 580t, 583 Antiseptics, for skin preparation, 23 Automated coagulometers, 784, 785t, 788-789, 793t.
physiologic adaptations in, 242-243 Antithrombin, 639t, 640-641 See also Coagulation instrumentation.
red blood cell distribution width in, 243, 249, 250t deficiency of, thrombosis risk and, 671t-672t, 672, Autophagocytosis, 419-420
red blood cell histogram in, 243 676-677, 677f Autoradiography, of amplified DNA, 478, 478f
red blood cell indices in, 243, 244t in disseminated intravascular coagulation, 687t Autosplenectomy, 72-73
reference ranges for, 242 heparin resistance and, 678 Avascular necrosis, in sickle cell anemia, 375
refractory reference ranges for, 677 AVOX 1000E, 189
with excess blasts, 538t, 539 Antithrombin activity assay, 677, 677f Azacitidine, for myelodysplastic syndromes, 541
with ringed sideroblasts, 538t, 539, 539f Antithrombin antigen assay, 677-678 Azathioprine, for warm autoimmune hemolytic
reticulocyte count in, 243-244, 244t, 248, 248f Antithrombotic therapy. See also Anticoagulants. anemia, 357
sickle cell. See Sickle cell anemia. monitoring of, 765-782 Azithromycin-atovaquone, for babesiosis, 347-348
sideroblastic, 260-261, 260b, 260f Antithymocyte globulin with cyclosporine, for aplastic Azure granules
in lead poisoning, 260-261 anemia, 288 in monocytes, 144-145, 145f
from porphyrias, 261, 261t Antitumor antibiotics, for leukocyte neoplasms, 430, in neutrophils, 136-137, 136b, 137f
spur cell, 324 431t
in thalassemia, 393-394 Aperture diaphragm, 35, 36f
in -thalassemia intermedia, 397 Apixaban, 774-775 B
in -thalassemia major, 395 Aplastic anemia, 284-291, 284b B cell(s)
Anesthetics acquired, 284-288 development of, 146-147, 147f
for bone marrow biopsy, 213-214 clinical findings in, 287 differentiation of, 559-560, 560f
platelet dysfunction from, 726 etiology of, 284-285, 285b in elderly, 588-589
Aneuploidy, 452 incidence of, 284 flow cytometry markers of, 491t, 495, 559-560,
Angioblasts, 66 laboratory findings in, 287-288, 287f-288f, 287t 560f
Angular cheilosis, in iron deficiency anemia, 256 treatment and prognosis for, 288 functions of, 148
Anion exchanger 1 (band 3), 108t classification of, 284b genetic abnormalities of, 415-416
in hereditary ovalocytosis, 322-323 differential diagnosis of, 290-291, 291t-292t germinal center, 560, 560f
in hereditary spherocytosis, 316 in dyskeratosis congenita, 289-290 immature, 147, 147f
Anisocytosis, 245t in Fanconi anemia, 288-289 memory, 560
in neutrophils, 420, 420f inherited, 288-290 reactive alterations in, 421-422
in sickle cell anemia, 375-376, 376f in Shwachman-Diamond syndrome, 290 B-cell lymphoma, 563-573, 565t
Ankyrin, 108, 109f, 110, 111t Aplastic crisis, in hereditary spherocytosis, 320 diffuse large, 565t, 571-572, 572f
in hereditary spherocytosis, 316 Apoferritin, 130 flow cytometry in, 502, 503f
Annular diaphragm, 38 Apoptosis, 63-64, 63f, 78 morphologic and immunophenotypic features of,
Antecubital fossa veins, for venipuncture, 23, 24f disruption of, in myelodysplastic syndromes, 534 565t
Anti-P autoantibody, in paroxysmal cold inhibition of, erythropoietin-induced, 96-97 B-cell prolymphocytic leukemia, 565t, 566-567, 566f
hemoglobinuria, 359 in leukocyte neoplasms, 429 B lymphoblastic leukemia-lymphoma
Antibiotics Apotransferrin, 129 flow cytometry in, 500-501, 501f, 547-548
antitumor, for leukocyte neoplasms, 430, 431t APTT. See Activated partial thromboplastin time genetics of, 548
beta-lactam, platelet dysfunction from, 725-726, 725f (APTT). Philadelphia chromosomepositive, 548
in sickle cell anemia, 377-378 Aquaporin 1, 108t, 111 subtypes of, 547, 547b
Antibody(ies) Ara-C, 431t Babesiosis, 347-348, 347f-348f
antiphospholipid Arachidonic acid Bacteria
assays for, 673, 674f-675f disorders of, 723 in body fluids, 233, 233t
drug-induced, 672, 673b in platelet activation, 166-167, 630, 631f in cerebrospinal fluid, 229-230, 230f
thrombosis risk and, 672-675 in platelet aggregometry, 743t, 745, 746t in phagocytic vacuoles, 420, 420f
Index 845
Bacterial infection Blood group Bone marrow specimens
in sickle cell anemia, 375 ABO anticoagulated aspirate smears of, 215
thrombocytopenia from, 697 accidental transfusion of, hemolytic reaction to, buffy coat smears of, 215
Band 3 (anion exchanger 1), 108t 361 collection procedures for, 214
in hereditary ovalocytosis, 322-323 von Willebrand factor reference intervals for, 658, collection sites for, 212, 212f
in hereditary spherocytosis, 316 658t crush smears of, 215
Band neutrophils, 2f, 3, 138, 139f hemolytic disease of fetus and newborn and, direct aspirate smears of, 214-215
Bartonellosis, 348 361-362, 362t, 706 dyes/stains for, 215, 216t, 219, 220t
Basement membrane, 69 Blood group antigens, 108, 109f histologic sections (cell block) of, 215
Basilic vein, for venipuncture, 23, 24f Blood smear, peripheral. See Peripheral blood smear. imprints (touch preparations) of, 215
Basopenia, 417 Bloodborne pathogens, standard precautions for, 8-12, management of, 214-215
Basophil(s), 2f, 3, 134, 142-144 9f-12f Bone marrow transplantation
development of, 142-143, 143f Body cavity effusions, in primary myelofibrosis, 524 for aplastic anemia, 288
functions of, 143-144 Body fluids, 226-240 for chronic myelogenous leukemia, 515-517
granules of, 144b automated counting of, 617 for Diamond-Blackfan anemia, 292-293
kinetics of, 143 in bronchoalveolar lavage specimens, 237-238, for dyskeratosis congenita, 290
in neonates, 585 238f for Fanconi anemia, 289
in serous fluid, 232 case study on, 226 for sickle cell anemia, 378
Basophil count cell counts on, 227, 228t for -thalassemia major, 396-397
abnormal, 417 cerebrospinal, 228-231, 229f-231f, 232t. See also Bottle carriers, 13
automated, 602t Cerebrospinal fluid. Breast feeding
Basophil-lobularity (BASO) channel, in Siemens cytocentrifuge slide preparation of, 227-228, 228f folate and vitamin B12 deficiency and, 273
hematology analyzers, 611-612, 613f Gram-stained organisms in, 233, 233t iron deficiency and, 253-254, 256
Basophilia, 3, 417 serous, 232-234, 232t, 233f-235f Brill-Edwards curve, 47
diffuse versus punctate, 94b, 246t standard precautions for, 8-12, 9f-12f Bronchoalveolar lavage specimens
persistence of, in myelodysplastic syndromes, synovial, 234-236, 236f-237f differential cell counts of, 237-238, 238f
535-536, 535f Body temperature, oxygen dissociation curve and, 122 procedure and precautions for, 237
Basophilic normoblast, 87, 90f-91f, 90t, 91 Bohr effect, 121, 121f Bruise, 648
Basophilic stippling, 94b, 246t, 247f Bone changes, in -thalassemia major, 395, 396f during specimen collection, 25
in sideroblastic anemia, 260-261 Bone marrow, 67-68 in thrombocytopenia, 695
in -thalassemia major, 395, 396f anatomy and architecture of, 211 Bubbles, in immersion oil, 37
in -thalassemia minor, 395, 395f cell counts and distributions in, 216-217, 218t, Buffy coat, 2, 179f
BCR-ABL1 fusion gene 219-220, 221f quantitative analysis of, 189
ATP binding site mutations of, 515-516, 517f circulation in, 69-70 Buffy coat smear
in B lymphoblastic leukemia-lymphoma, 493, hematopoiesis in, 62-63 of bone marrow specimens, 215
500-501 fetal, 67 myeloid-erythroid layer in, 215
in chronic myelogenous leukemia, 455-456, 456f hypoplasia of, bone marrow failure from, 292t Burkitt lymphoma, 429, 565t, 572-573, 572f
molecular biology of, 510, 511f lymphocyte development in, 135, 147 Burn injury, hemolytic anemia from, 349, 349f
signaling pathways influenced by, 510-512, 512f panmyelosis of, in polycythemia vera, 518, 520, Burr cells, 245t, 247f, 324
increased copy number of, 515-516, 516f 521f in pyruvate kinase deficiency, 332
real-time polymerase chain reaction analysis of, red marrow of, 68-69, 68f-70f Burst-forming uniterythroid (BFU-E), 79, 87
483, 515 Bone marrow aspiration Burst-forming unitmegakaryocyte (BFU-Meg), 153,
reverse transcription polymerase chain reaction collection procedures for, 214 154f, 155t
amplification of, 476, 476f-477f examination of aspirate smears or imprints after Busulfan, 431t
BCR1 gene, 510-511 high-power (500x), 216b, 217-219, 218f-219f, Butterflies (winged infusion sets), 20-22, 22f
BCS coagulometers, 793t 218t for hemostasis specimen collection, 735-736,
Becton Dickinson Eclipse needle, 20 low-power (100x), 216-217, 216b, 216f-217f 737f
Bee stings, hemolytic anemia after, 348-349 using Prussian blue iron stain, 219, 219f Bypass surgery
Benign cells, characteristics of, 232t needles for, 213-214 platelet disorders and, 727
Benzalkonium chloride, skin cleansing with, 23 Bone marrow biopsy thrombocytopenia and, 709
Bernard-Soulier syndrome, 719-720, 720f collection procedures for, 214
Bethesda assay, for factor VIII inhibitors, 661 examination of specimen after, 219-220, 220f-222f,
Bias, 44 220t C
Bicarbonate, 122 indications for, 212 C-banding stain, 449, 450f
Bilirubin needles for, 213, 213f-214f C-kit ligand, 79, 81t-82t
conjugated, 301-302 Bone marrow examination, 5, 210-225 C-reactive protein, high-sensitivity, arterial thrombosis
in hemolytic process, 307-308, 308b in anemia, 244-246 risk and, 680-681, 680t-681t
indirect serum, in thalassemia, 403 in aplastic anemia, 287, 288f Cabot ring, 246t
laboratory testing for, 308b blood smear role in, 212-213 Calcium
in megaloblastic anemia, 276 case study on, 210 in coagulation, 632t, 633
metabolism of, 300f, 301-303 in chronic eosinophilic leukemia, not otherwise deficiency of, in acute myeloid leukemia, 549
unconjugated, 301-302 specified, 526-527 intracellular concentration of, 112
Bilirubin diglucuronide, 300f, 301-302 in chronic lymphocytic leukemia/small lymphocytic mobilization defects of, 723-724
Bilirubinemia, 309-310, 310t lymphoma, 563, 566f in platelets, 162t
Biliverdin, 300f, 301-302 in chronic myelogenous leukemia, 512, 513t, 514f Calcium ATPase, 108t, 112
Biohazard waste in chronic neutrophilic leukemia, 526 Calcium pyrophosphate crystals, in synovial fluid,
containers for, 19 definitive studies in, 220-221, 222t 236, 236f
symbol for, 9f, 19 in essential thrombocythemia, 522-523, 522f, 522t Calibration verification, 47-48
Biologic response modifiers, thrombocytopenia from, in hairy cell leukemia, 567, 567f Calibrators, 42-43, 47-48
705-706 in hemolytic process, 309 Canalicular system, surface-connected, platelet, 158
Biopsy. See Bone marrow biopsy; Lymph node indications for, 211-212, 211t Cancer. See also Neoplasms.
biopsy. in iron deficiency anemia, 258, 258f cytogenetic analysis in, 453-458, 455t
Biotin, as hybridization label, 480, 481f in megaloblastic anemia, 277, 277f disseminated intravascular coagulation and, 683
Birth weight, hematologic values and, 581 in myelodysplastic syndromes, 534-537, 538t thrombocytopenia in, 698
2,3-Bisphosphoglycerate, 105f, 106-107 in paroxysmal nocturnal hemoglobinuria, 327 thrombosis risk in, 670, 671t
in anemia, 242-243 patient care after, 214 Capillary blood
oxygen affinity and, 121f-122f, 122 in polycythemia vera, 520, 520t, 521f for hemostasis specimens, 738
Bite cells, in glucose-6-phosphate dehydrogenase preparation for, 212-214, 213f-214f skin puncture for, 27-28, 27f-28f
deficiency, 330-331 in primary myelofibrosis, 524 Capillary tubes, 28
Bivalirudin, 776 reports following, 221, 223f, 224t in hematocrit determination, 179f, 180
monitoring of, 776 sample for. See Bone marrow specimens. Carbaminohemoglobin, 122
prothrombin time and, 769 in sickle cell anemia, 375-376 Carbohydrates, membrane, 60
Blast cells in thalassemia, 401 Carbon dioxide, transport of, 122
excess, refractory anemia with, 538t, 539 Bone marrow failure, 283-298 Carbon monoxide, 123
flow cytometry markers of, 495-496, 496f in anemia of chronic kidney disease, 294 in hemolytic process, 308
Blast crisis, in chronic myelogenous leukemia, 513 in aplastic anemia, 284-291, 284b. See also Aplastic Carbonic anhydrase, 122
Blastic plasmacytic cell neoplasm, 554-555 anemia. Carboxyhemoglobin, 123, 178
Bleach disinfectant solution, 11 case study on, 283 Cardiac catheterization, glycoprotein IIb/IIIa
Bleeding. See Hemorrhage. clinical consequences of, 284 antagonists during, 776-777
Bleeding time test, 50, 741 in congenital dyserythropoietic anemia, 293, 293f Cardiomegaly, in sickle cell anemia, 375
Bleomycin, 431t inherited/congenital, key characteristics of, 292t Cardiopulmonary bypass surgery
Blind loops, megaloblastic anemia from, 275 in myelophthisic anemia, 294 platelet disorders and, 727
Blood in paroxysmal nocturnal hemoglobinuria, 327, thrombocytopenia and, 709
collection of. See Specimen collection. 328t Carmustine, 431t
loss of. See Hemorrhage. pathophysiology of, 284 Carrin disease, 348
transfusion of. See Transfusions. in pure red cell aplasia, 291-293 Cartilage cells, in cerebrospinal fluid, 230, 231f
Blood culture, specimen collection for, 29 in sickle cell anemia, 375 Cascade POC Actalyke, 792t
846 Index

Catheterization Cell cycle, 63 Chromogenic end-point detection, 783t, 786, 787f


cardiac, glycoprotein IIb/IIIa antagonists during, of chromosomes, 446, 447f Chromogenic factor X assay, 786, 787f
776-777 DNA replication and, 467-470, 469f-473f for warfarin monitoring, 768
central venous, thrombosis risk and, 670t regulation of, 63, 470, 472f Chromosomes
Cation pumps, ATP-dependent, 111 Center for Medicare and Medicaid Services (CMS), 53 abnormalities of, 452-453
CBC. See Complete blood count. Central dogma in genetics, 463-464, 464f in cancer, 453-458, 455t
CD antigens Central venous catheterization, thrombosis risk and, in leukemia, 455-457, 455b, 455t, 456f-457f
hematolymphoid, function and cellular expression 670t in leukocyte neoplasms, 428
of, 491-492, 492t Centrifuge systems, 180, 180f, 188 in myelodysplastic syndromes, 540
lineage-associated, 491, 491t, 494-495 double-spin approach in, 740 numeric, 452, 452f
CD1a, 492t swinging bucket, 740 in solid tumors, 455t, 457-458, 457f
CD2, 492t Centrioles, 62 structural, 452-453, 453f-454f
CD3, 492t Centroblast, 560 analysis of, 445-460. See also Cytogenetic analysis.
CD4, 148, 492t Centrocyte, 560 cell cycle of, 446, 447f
CD5, 492t Cephalic vein, for venipuncture, 23, 24f metaphase, 447
CD7, 492t Cerebrospinal fluid, 228-231, 229f number of, 447
in acute myeloid leukemia, 496, 498f bacteria in, 233t Philadelphia. See Philadelphia chromosome.
CD8, 148, 492t cell counts of, 229 ring, 453, 454f
CD10, 492t differential cell counts of, 229-231, 230f-231f, 232t size and type of, 447-448, 448f
CD11b, 492t gross examination of, 228-229, 229t structure of, 446-447, 447f-448f
CD13, 492t Certification, medical laboratory, 51-53 Chronic eosinophilic leukemia, not otherwise
CD14, 492t Cesarean section, in neonatal alloimmune specified, 526-527
CD15, 492t thrombocytopenia, 704-705 Chronic leukemia, 3
in Hodgkins lymphoma, 576 Chdiak-Higashi syndrome, 161-162, 410, 411f, 722 Chronic lymphocytic leukemia, 563-566, 563f, 565t,
CD16, 492t Cheilosis, angular, in iron deficiency anemia, 256 566f
CD18, 492t Chemical hazards, 13-14 Chronic monocytic leukemia, 513
CD19, 492t Chemicals, flammable or combustible, storage of, 13 Chronic myelogenous leukemia, 509-517
CD20, 492t Chemistry studies, in anemia, 246 atypical (BCR/ALB1 negative), 540
CD22, 492t Chemotherapy BCR-ALB1 fusion gene in, 455-456, 456f
CD30 for acute lymphoblastic leukemia, 548-549 molecular biology of, 510, 511f
in anaplastic large cell lymphoma, 574-575 folate in, 272b signaling pathways influenced by, 510-512, 512f
in Hodgkins lymphoma, 576 for leukocyte neoplasms, 430, 431t chromosomal abnormalities in, 455-456, 456f
CD31 (PECAM), 160, 492t leukocyte neoplasms from, 427-428 flow cytometry in, 499-500
CD32, 160 thrombocytopenia from, 697 imatinib for, 432, 515-517, 515f
CD33, 492t Chest, acute, in sickle cell anemia, 374 resistance to, 515-516, 516f-517f
CD34, 492t Children, 581-586. See also Infants; Neonates. incidence of, 510
in aplastic anemia, 285-286 anemia in, 584 versus leukemoid reaction, 416, 416b
in primary myelofibrosis, 523-524 cytopenia in, refractory, 539 molecular genetics of, 510, 511f
CD36, 492t erythroblastopenia in, transient, 291-292 oncogenes in, 429
CD38, 492t hematologic values in other laboratory findings in, 512
CD41, 492t developmental stages of, 581 pathogenesis of, 510-512, 512f
CD42b, 492t by gestational age and birth weight, 581 peripheral blood and bone marrow in, 512, 513t,
CD45, 492t for platelets at birth, 586, 586t 514f
in blast cells, 495-496, 496f for red blood cells at birth, 581-584, 582f Philadelphia chromosome in, 510
in flow cytometry, 494, 494f for very low birth weight infants during first 6 progression of, 513
CD50, 160 weeks, 583, 583t related diseases in, 513-514
CD54, 160 for white blood cells at birth, 584-586, 584t-585t, treatment of, 514-517
CD55, in paroxysmal nocturnal hemoglobinuria, 585f Chronic myeloid neoplasms, in elderly, 591
325-327, 327f hemolytic uremic syndrome in, 708 Chronic myelomonocytic leukemia, 513, 540
CD56, 492t hemostasis in, 586, 587t-588t Chronic neutrophilic leukemia, 513, 526
CD59, in paroxysmal nocturnal hemoglobinuria, Henoch-Schnlein purpura in, 729 Chylomicrons, hematology analyzer interference due
325-327, 327f, 504f, 505 immune thrombocytopenic purpura in, 698-700, to, 616t
CD61, 492t 700t Ciliated epithelial cells, in bronchoalveolar lavage
CD62, 160, 629 iron deficiency anemia in, 584 specimens, 237-238, 238f
CD62P, 492t myelodysplastic syndromes in, 539 Circulation, in bone marrow, 69-70
CD63, 492t needle gauges for, 25 Cirrhosis, alcoholic, platelet disorders in, 727
CD64, 492t sepsis in, 585-586 Cisplatin, 431t
CD71, 492t venipuncture in, 25 Cisternae, 60
CD79a, 492t Chlorambucil, 431t Citrate, in collection tubes, 20
CD102, 160 Chlorhexidine gluconate/isopropyl alcohol, skin Citrate theophylline adenosine dipyridamole, for
CD117, 492t cleansing with, 23 hemostasis specimens, 739
2-CDA, 431t Chloride-potassium cotransporter, 108t Clindamycin, for babesiosis, 347-348
CDAN1 gene, in congenital dyserythropoietic anemia, Chloride-potassium-sodium cotransporter, 108t Clinical and Laboratory Standards Institute (CLSI),
293 Chloride-sodium cotransporter, 108t 53
Celiac disease, folate absorption in, 273 Chloroquine venipuncture steps recommended by, 23-25
Cell(s), 57-65 for malaria, 346-347 Clinical efficacy, 50-51, 50t-51t
components and functions of, 59t resistance to, 346-347 Clinical laboratory hematology. See Laboratory
cytoplasm of, 60-62, 61f Chlorothiazide diuretics, thrombocytopenia from, 697 hematology.
death of, 63-64, 63f Choking, during specimen collection, 26 Clinical laboratory science degree, 53
membrane of, 57-60 Cholelithiasis Clonality, in lymphoblastic leukemia-lymphoma, 500
nucleus of, 60 in hemolysis, 306 Clone, 454-455
organization of, 57, 58f in hereditary elliptocytosis, 322 Clopidogrel
programmed death of. See Apoptosis. in hereditary spherocytosis, 320 antiplatelet activity of, 777, 777f
Cell block, of bone marrow specimens, 215 Cholesterol, 59 monitoring of, platelet aggregometry for, 746
Cell counts in platelet plasma membrane, 158, 158f platelet dysfunction from, 725
automated, 598-625. See also Hematology analyzers. in red blood cell membrane, 107-108 for prophylaxis, 778
body fluid, 227, 228t total response variation to, 778
bone marrow, 216-217, 218t, 219-220, 221f fibrinogen and, 682, 683f thrombotic thrombocytopenic purpura from,
cerebrospinal fluid, 229 thrombosis risk and, 680, 680t 706-707
cytocentrifuge slide preparation in, 227-228, 228f Cholesterol crystals, in synovial fluid, 236 Clostridial sepsis, 348
differential Cholesterol:high-density lipoprotein ratio, 680, 680t Clot
in body fluid analysis, 227-228 Chorea acanthocytosis, 324-325 formation of. See Coagulation.
in bone marrow examination, 218 Choroid plexus cells, in cerebrospinal fluid, 230 red, 163-164
of bronchoalveolar lavage specimens, 237-238, Christmas disease, 662, 662f white, 163, 166f
238f Chromatic aberrations, in microscopy, 33-35 Clot activator, in collection tubes, 20
of cerebrospinal fluid, 229-231, 230f-231f, 232t Chromatin, 60, 447 Clot-based studies. See also Coagulation testing.
of serous fluid, 232-234, 233f-235f Chromatography, high-performance liquid, 403 for activated protein C resistance, 676, 676f
of synovial fluid, 234-236, 236f for homocysteine, 681, 682t in congenital single-factor deficiencies, 660, 660t
of white blood cells. See White blood cell Chromium labeling, in hemolytic process, 308 in hemophilia, acquired, 654, 654f
differential. Chromogenic antifactor Xa heparin assay instruments for. See Coagulation instrumentation.
manual, 173-174 for fondaparinux monitoring, 774 for lupus anticoagulant, 674, 752-753
calculations for, 173 for heparin monitoring, 773-774 partial thromboplastin time in, 750-752, 751f
equipment for, 173, 174f for rivaroxaban monitoring, 774-775 partial thromboplastin time mixing studies in,
of platelets, 175, 176t Chromogenic assay 752-753
of red blood cells, 176, 176t for antithrombin, 677 platelet-poor plasma specimens for, 740-741
of white blood cells, 174, 174b, 174f-175f, 176t for plasminogen, 758-759, 759f for procoagulant screens, 747-754
point-of-care testing of, 189 for protein C, 679, 679f for protein C, 679
Index 847
Clot-based studies (Continued) Coagulopathy (Continued) Coulter hematology analyzers, 2, 599f, 602-604, 602t,
for protein S, 679, 679f symptoms of, 647-649, 648b 604f-605f
prothrombin time in, 748-750, 748f, 750t in vitamin K deficiency, 652-653, 653f VCS technology in, 603-604
reptilase time in, 753f, 754 in von Willebrand disease. See also Von Willebrand COULTER LH slide maker and stainer, 197f
thrombin time in, 753-754, 753f disease. Coumarins, 766-767. See also Warfarin.
Clothing, personal protective, 9-10 acquired, 654-655 Coverslip
Clotting time congenital, 655-663, 655f-656f, 655t, 657t-659t in bone marrow sample management, 215
activated, 747 COAT platelets, 638 in peripheral blood smear, 194, 196f
for direct thrombin inhibitor therapy monitoring, CoatLab coagulometers, 793t Cranial bleeds
776 Coats, laboratory, 9-11 in hemophilia A, 659
for unfractionated heparin monitoring, 772 Cobalamin. See Vitamin B12 (cobalamin). in immune thrombocytopenic purpura, 698
Ecarin, for direct thrombin inhibitor therapy Coefficient of variation in percent (CV%), 45 Creatinine assay, during low-molecular-weight heparin
monitoring, 776 Cofactors, procoagulant, 633, 633t therapy, 773
kaolin, for lupus anticoagulant detection, 673-674, Cold agglutinins, 181, 357-359, 357t, 358f Cristae mitochondria, 61-62
674f hematology analyzer interference due to, 616-617, Critical illumination, 35
Cluster of differentiation (CD). See CD entries. 616t Crush smears, of bone marrow specimens, 215
CML. See Chronic myelogenous leukemia. Cold hemoglobinuria, paroxysmal, 357t, 359 Cryohemolysis test, hypertonic, in hereditary
CoaguChek XS and XS Plus, 792t Collagen, 70 spherocytosis, 320
Coagulation, 163-165, 627, 627f, 631-639 in hemostasis, 628-629 Cryoprecipitate
cell-based (in vivo), 637-639, 638f in platelet aggregometry, 743t, 745, 746t for acute coagulopathy of trauma-shock, 650
clot formation graph of, 789, 789f in primary myelofibrosis, 523 for disseminated intravascular coagulation,
contact factors in, 637 Collection tubes 686-687
disseminated intravascular, 655, 683-687 additives in, 20 for gray platelet syndrome, 723
activated partial thromboplastin time in, 686, for hemostasis specimens, 735 for liver diseaserelated hemorrhage, 652
686t, 752 labeling of, 24 Crystals
etiology of, 683, 684t micro-, 28 Hb C, 245t, 247f
hemolytic anemia in, 341 for skin puncture, 27f, 28 Hb SC disease, 245t, 247f, 381, 382f
laboratory evaluation of, 686, 686t-687t for venipuncture, 20, 21f in synovial fluid, 236, 236f
in liver disease, 651 College of American Pathologists (CAP), 53-54 Cyanmethemoglobin, 2, 123
pathophysiology of, 683-686, 685f external quality assessment by, 50 Cyanmethemoglobin method, for hemoglobin
platelet disorders in, 728 Q-PROBES program of, 53 determination, 177, 177f-178f
symptoms of, 686 Colloid osmotic hemolysis, 111 Cyanosis, case study on, 103-104
thrombocytopenia in, 709 Colony-forming uniterythroid (CFU-E), 87 Cyclin, 63
treatment of, 686-687 Colony-forming unitmegakaryocyte (CFU-Meg), 153, Cyclin-dependent kinases, 63
extrinsic, intrinsic, and common pathways in, 154f, 155t in leukocyte neoplasms, 429
637 Colony-forming units (CFUs), 76, 76t Cyclooxygenase pathway, in platelet activation,
factor XI activation pathway in, 637 Colony-stimulating factors, 79, 81t-82t 166-169, 168f
fibrinolysis in, 628, 641-644, 642f-643f, 642t for leukocyte neoplasms, 431-432 Cyclophosphamide, 431t
inhibitors of. See Anticoagulants. thrombocytopenia from, 705-706 for warm autoimmune hemolytic anemia, 357
procoagulants in Common lymphoid progenitor, 76 Cyclosporine, antithymocyte globulin with, for
biochemical nature of, 633-635, 633t, 634f-636f Common myeloid progenitor, 76 aplastic anemia, 288
nomenclature for, 631-633, 632f, 632t Comparative genomic hybridization, 458, 458f CYP2C9 polymorphisms, warfarin and, 768-769
physiologic function of, 633, 633b, 633t Competence, laboratory staff, 51-53 Cystathionine, 681, 682f
promotion of, vascular intima (endothelium) and, Complement, in immune hemolytic anemia, 355 Cytarabine, for chronic myelogenous leukemia,
629, 629t Complete blood count, 4-5, 192-209 515
regulation of, 639-641 in anemia, 243, 244t Cytocentrifuge slides, in body fluid analysis, 227-228,
protein C in, 639t, 640, 640f in aplastic anemia, 287, 287t 228f
serine protease inhibitors (serpins) in, 639t, automated, 602t Cytochemical staining, 6, 222t, 436-444. See also
640-641, 641f case study on, 192 Stain(s); Staining.
tissue factor pathway inhibitor in, 639-640, 639f, in hemoglobin SC disease, 381 acceptable specimens and fixatives for, 437
639t in hemoglobinemia, 303b case study on, 436
thrombin in, 637 in hemolytic process, 307, 308b, 309 controls and troubleshooting for, 442
tissue factor pathway in, 629, 635-637, 636f, 637t interpretation of, phases in, 202-203 interpretation of, 437-442, 440t
tissue factor pathway inhibitor in, 628-629 in iron deficiency anemia, 256f, 257 for leukocyte disorders, 436-444, 440t
Coagulation factors. See Factor entries; Procoagulants. in megaloblastic anemia, 276, 276f principles of, 436
Coagulation instrumentation, 783-795 parameters of, 4, 4b Cytochrome b5 reductase, 106
advances in, 787-789, 788b, 789f peripheral blood film in. See Peripheral blood Cytochrome oxidase iron, 127
advantages and disadvantages of, 789, 790t smear. Cytogenetic analysis, 6, 222t, 445-460
case study on, 783 results from in aplastic anemia, 287-288
currently available, 793, 793t organization of, 203

You might also like