You are on page 1of 23

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/5253256

Suggested Guidelines for Immunohistochemical


Techniques in Veterinary Diagnostic Laboratories

Article in Journal of veterinary diagnostic investigation: official publication of the American Association of Veterinary
Laboratory Diagnosticians, Inc · August 2008
DOI: 10.1177/104063870802000401 · Source: PubMed

CITATIONS READS

89 166

15 authors, including:

Matti Kiupel Stefanie Czub


Michigan State University Canadian Food Inspection Agency
307 PUBLICATIONS 4,450 CITATIONS 128 PUBLICATIONS 2,638 CITATIONS

SEE PROFILE SEE PROFILE

Sharon Dial Lisa Manning


The University of Arizona Health Sciences Centre Winnipeg
40 PUBLICATIONS 817 CITATIONS 8 PUBLICATIONS 369 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Wild and Exotic Animal Ophthalmology - Morphological Descriptions, Clinical and Surgical Procedures
View project

chronic wasting disease View project

All content following this page was uploaded by Matti Kiupel on 31 December 2013.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
Journal of Veterinary Diagnostic
Investigation http://vdi.sagepub.com/

Suggested Guidelines for Immunohistochemical Techniques in Veterinary Diagnostic Laboratories


José A. Ramos-Vara, Matti Kiupel, Timothy Baszler, Laura Bliven, Bruce Brodersen, Brian Chelack, Keith West, Stefanie
Czub, Fabio Del Piero, Sharon Dial, E. J. Ehrhart, Tanya Graham, Lisa Manning, Daniel Paulsen and Victor E. Valli
J VET Diagn Invest 2008 20: 393
DOI: 10.1177/104063870802000401

The online version of this article can be found at:


http://vdi.sagepub.com/content/20/4/393

Published by:

http://www.sagepublications.com

On behalf of:

Official Publication of the American Association of Veterinary Laboratory Diagnosticians, Inc.

Additional services and information for Journal of Veterinary Diagnostic Investigation can be found at:

Email Alerts: http://vdi.sagepub.com/cgi/alerts

Subscriptions: http://vdi.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

>> Version of Record - Jul 1, 2008

What is This?

Downloaded from vdi.sagepub.com by guest on October 11, 2013


J Vet Diagn Invest 20:393–413 (2008)

Suggested guidelines for immunohistochemical techniques in veterinary


diagnostic laboratories
José A. Ramos-Vara,1 Matti Kiupel,2 Timothy Baszler, Laura Bliven, Bruce Brodersen,
Brian Chelack, Stefanie Czub, Fabio Del Piero, Sharon Dial, E. J. Ehrhart, Tanya Graham,
Lisa Manning, Daniel Paulsen, Victor E. Valli, Keith West

Abstract. This document is the consensus of the American Association of Veterinary Laboratory
Diagnosticians (AAVLD) Subcommittee on Standardization of Immunohistochemistry on a set of guidelines
for immunohistochemistry (IHC) testing in veterinary laboratories. Immunohistochemistry is a powerful ancillary
methodology frequently used in many veterinary laboratories for both diagnostic and research purposes. However,
neither standardization nor validation of IHC tests has been completely achieved in veterinary medicine. This
document addresses both issues. Topics covered include antibody selection, fixation, antigen retrieval, antibody
incubation, antibody dilutions, tissue and reagent controls, buffers, and detection systems. The validation of an
IHC test is addressed for both infectious diseases and neoplastic processes. In addition, storage and handling of
IHC reagents, interpretation, quality control and assurance, and troubleshooting are also discussed. Proper
standardization and validation of IHC will improve the quality of diagnostics in veterinary laboratories.
Key words: Animal diseases; diagnostic immunohistochemistry; standardization; validation.

Introduction istry (IHC) has been proven to be one of the most


important ancillary techniques in the characterization
Morphological diagnosis in veterinary medicine has
of neoplastic diseases in humans and has become
classically relied mostly on routine stains such as
equally important in veterinary medicine, as oncolo-
hematoxylin and eosin and, less commonly, on other
gists demand more specific diagnoses. The number of
histochemical stains. However, the level of specializa-
immunohistochemical tests offered by veterinary diag-
tion in veterinary practice demands more accurate
nostic laboratories for the diagnosis of infectious and
diagnosis, particularly for tumors. Immunohistochem- neoplastic diseases in frozen or formalin-fixed, paraf-
fin-embedded (FFPE) tissues has increased exponen-
From the Animal Disease Diagnostic Laboratory, Purdue
University, West Lafayette, IN (Ramos-Vara), Diagnostic Center tially in the last decade. Immunohistochemistry has
for Population and Animal Health, Michigan State University, East been proven as a highly specific and sensitive diagnostic
Lansing, MI (Kiupel), Washington Animal Disease Diagnostic method that is especially advantageous as a diagnostic
Laboratory, Washington State University, Pullman, WA (Baszler), tool for infectious diseases. In some cases, IHC is
Marshfield Clinic Laboratories Veterinary Services, Marshfield, WI
considered the gold standard technique to which others
(Bliven), Veterinary Diagnostic Laboratory, University of Ne-
braska, Lincoln, NE (Brodersen), Prairie Diagnostic Services Inc., are compared (e.g., prion diseases). In comparison with
Saskatoon, Saskatchewan, Canada (Chelack, West), National other diagnostic tests, IHC allows colocalization of an
Centre for Foreign Animal Diseases, Canadian Food Inspection antigen with a lesion, thereby dramatically increasing
Agency, Winnipeg, Canada (Czub), Department of Pathobiology diagnostic accuracy and understanding of pathogene-
and Clinical Studies, New Bolton Center, University of Pennsylva-
nia, Kennett Square, PA (Del Piero), Department of Veterinary
sis. Numerous infectious agents and cell types can be
Science and Microbiology, University of Arizona, Tucson, AZ identified with IHC in a wide variety of animal species,
(Dial), Department of Microbiology, Immunology, and Pathology, especially since the advent of antigen retrieval methods
Colorado State University, Fort Collins, CO (Ehrhart), Animal using heat.62 However, there are no guidelines for
Disease Research and Diagnostic Laboratory, South Dakota State standardization and no general agreement on the use of
University, Brookings, SD (Graham), National Centre for Foreign
Animal Diseases, Canadian Food Inspection Agency (Manning),
controls or the validation of a test in veterinary
Department of Pathobiological Sciences, Louisiana State Univer- diagnostic IHC. The purpose of the present article is
sity, Baton Rouge, LA (Paulsen), and Department of Pathobiology, to provide guidelines for IHC in the veterinary
University of Illinois, Champaign, IL (Valli). diagnostic laboratory, and is not intended as manda-
1
Corresponding Author: José A. Ramos-Vara, Animal Disease tory requirements for laboratory accreditation. The
Diagnostic Laboratory, Purdue University, 406 South University,
West Lafayette, IN 47907. ramosja@purdue.edu article briefly reviews the technical aspects of IHC and
2
Dr. Kiupel is Chair of the AAVLD Subcommittee on provides guidance for standardization, test validation,
Standardization of IHC. controls, storage and handling, interpretation and
393
394 Ramos-Vara et al.

Figure 1. Overview of immunohistochemical test. The immunohistochemistry technique can be divided into 4 main sections:
pretreatments (stage 1), immunologic and histochemical reactions (stage 2), visualization of the immunologic reaction (stage 3), and
interpretation and report generation (stage 4).

quality control and assurance, as well as general tions, controls, buffers, detection system) to ensure
troubleshooting protocols. that the selected antibody reacts with the expected
antigen (Fig. 2). There is a lack of standardization
Overview of the immunohistochemical test among different veterinary laboratories, despite the
The IHC technique is a combination of immuno- widespread use of IHC.47 Contrary to the situation in
logic and chemical reactions visualized with a photonic human IHC, a major problem in veterinary IHC is
microscope. The IHC technique can be divided into 4 the reduced availability of high-quality antibodies
main steps (Fig. 1). Step 1 includes all preimmunologic that are specific for infectious agents of veterinary
procedures done before incubation with the primary importance, and cell markers specific for small and
antibody. These procedures include deparaffinization large animal species. Especially in tumor diagnostics,
and hydration of tissue sections, blocking nonspecific most laboratories use batteries of antibodies devel-
bindings, blocking endogenous activities (e.g., perox- oped for human tissues that are often insufficiently
idase, phosphatase, biotin), and antigen retrieval. Step validated for animal species. With the exception of
2 includes all the immunologic reactions between the the surveillance program for prion diseases (e.g.,
primary antibody and tissue antigens, the primary scrapie, chronic wasting disease), neither the proto-
antibody and secondary antibody and some additional cols nor the primary antibodies are uniform among
chemical reactions necessary to bind the reporter diagnostic laboratories.
molecule (label) such as peroxidase or alkaline There is also little guidance or regulation for the
phosphatase to the preformed immune complex. Step actual standardization of specific IHC tests in human
3 includes all the procedures necessary to visualize the medicine although a consensus on guidelines for
antigen-antibody binding. In IHC, this is achieved with quality assurance for IHC has been published by the
a chemical reaction in which the label reacts with its Clinical and Laboratory Standards Institute (for-
substrate and a chromogen to produce a colored merly National Committee on Clinical Laboratory
reaction product. Then, immunolabeled tissue sections Standards, 1999).1 The Food and Drug Administra-
are counterstained and coverslipped. Step 4 includes tion (FDA) has classified most IHC reagents and kits
interpretation and reporting of the IHC results. as ‘‘Analyte Specific Reagents,’’ thereby exempting
them from premarket notification (FDA regulations
Standardization of a new test
classify devices depending upon the degree of
Standardization determines optimal conditions regulation necessary to provide reasonable assurance
(e.g., incubation time, incubation temperature, dilu- of their safety and effectiveness). This ruling was
Immunohistochemical techniques in veterinary diagnostic laboratories 395

Figure 2. Steps to consider during the standardization of a new immunohistochemical test. Five main factors are involved in
immunohistochemistry standardization: animal tissue, fixation, primary antibody, antigen retrieval, and detection systems.

based on the assumption that most IHC tests are part Polyclonal antibodies (pAb) contain antibodies to a
of the pathologist’s report and, as such, will not be range of antigens and thus may cause greater
reported independently. For antibodies used as stand- nonspecific background staining and be less specific
alone tests in human pathology, premarket notifica- than mAb. Subjecting pAb to multiple adsorption
tion and specific FDA approval are required. For protocols for a variety of antigens will increase their
stand-alone tests in veterinary IHC, premarket specificity. For both mAb and pAb, the diagnostic
notification by regulatory agencies (U.S. Department specificity of an antibody detecting infectious agents
of Agriculture [USDA]) is not required. However, for is the positive immunoreactivity against the targeted
certain animal diseases (e.g., scrapie, chronic wasting agent only and lack of immunostaining in tissues
disease, Bovine viral diarrhea virus), IHC results are infected with other agents. The diagnostic specificity
commonly reported as stand-alone tests, so standard- of an antibody detecting cellular/tumor markers is the
ization of these tests among various laboratories is expected presence or absence of immunostaining in
urgently needed. The main goal of standardization in certain cell types, tissues, or tumors in multitissue
IHC is to obtain reproducible and consistent results control blocks (see Controls section).64 Analytical
within each laboratory and comparable results among sensitivity is defined as weak (positive) staining in the
different laboratories even when using different presence of the least amount of antigen. Polyclonal
procedures. In this review, each of the main antibodies may be more sensitive than mAb because
components of an IHC test, including primary they can bind several different epitopes on a single
antibody, fixation, antigen retrieval, pretreatments, antigen.64 Diagnostic sensitivity, defined as the
and detection systems, are addressed. proportion of known positive samples that test
positive,65 is established by comparing the test
Primary antibody characterization results on FFPE tissue with results using another
Monoclonal and polyclonal antibodies: sensitivity antibody that has been validated for the same analyte
and specificity. Monoclonal antibodies (mAb) or using a non-IHC method such as culture or
derived from a single B-cell clone and produced by polymerase chain reaction (PCR).
hybridoma technique provide excellent specificity Cellular markers.—With the exception of CD (clu-
because the antibody binds to a single epitope on 1 ster of differentiation) markers, there are few cellular
antigen.43 most mAb are of mouse origin, although antigens in animals for which species-specific
rabbit mAb are also commercially available.56,69 antibodies have been developed, and even fewer are
396 Ramos-Vara et al.

detectable in FFPE tissues. Most antibodies to destabilizing hydrophobic and hydrophilic bonding
cellular antigens used in veterinary IHC laboratories (hydrophobic areas are released from the repulsion of
have been developed against human or rodent water, and occupy a greater area). This causes a
antigens, and characterization data for their disruption of tertiary protein structure and results in a
specified use should be available. Cellular markers partially reversed (‘‘less folded’’) protein structure.43
should demonstrate reactivity with the appropriate The disruption of tertiary structures also changes
molecular weight antigen in Western blots (WB); physical properties, and proteins that are normally
however, WB immunoreactivity does not necessarily soluble in aqueous solution become insoluble.
imply or predict immunoreactivity in FFPE tissues. Coagulative fixation maintains tissue structure at
Interpretation of IHC results requires familiarity the light microscopic level fairly well, but results in
with the expected pattern of immunoreactivity based cytoplasmic flocculation as well as poor preservation
on location of the antigen (e.g., nuclear, membrane, of mitochondria and secretory granules.43 The most
or cytoplasmic staining; Fig. 3).76 For example, common types of coagulative fixatives are dehydrants
staining for cytokeratins and vimentin should be (alcohols and acetone) and strong acids (picric acid,
cytoplasmic, not nuclear; staining for thyroid tran- trichloroacetic acid).
scription factor 1 (TTF 1) in lung or thyroid tumors is Cross-linking fixation. Cross-linking fixatives
nuclear, not cytoplasmic.48,51 In other words, the form cross-links within and between proteins, within
presence of staining does not always indicate a and between nucleic acids, and between nucleic acids
positive reaction. In most cases, the degree of specific and proteins. The most common cross-linking
staining should vary between cells and in some cases fixatives are aldehydes (formaldehyde,
within different cell compartments; such cellular glutaraldehyde, chloral hydrate, glyoxal), with
distribution may have prognostic significance in neutral buffered formaldehyde most frequently used
certain tumors.33 in routine histopathology. Formalin fixation
Infectious agents.—For viral agents, positive stain- produces methylene bridges between free amino
ing should be detected in the appropriate tissues, cell groups and other functional groups of proteins,
types, and cellular location (if known), in association resulting in conformational changes in the tertiary and
with typical lesions. For protozoal and bacterial quaternary structures within proteins and cross-linking
pathogens, the morphology and location of the between tissue proteins, thus modifying the epitopes
stained organisms can provide some additional recognized by antibodies.3,15,21,22,43,71 However, it needs
information. to be emphasized that adequate fixation is essential in
Fixation IHC. Underfixation of tissues for IHC testing is one of
the major problems in human pathology.15 Tissues that
The ultimate goal of fixation is to preserve cells and are poorly fixed in formalin will be subjected
tissues, to prevent autolysis, and to preserve antige- to coagulative fixation (alcohol) during tissue
nicity. Unfortunately, no fixative can optimally fulfill processing, resulting in inadequate antigen retrieval or
all these requirements.43 The most common type of unexpected antigen detection. Underfixed tissues
fixation in diagnostic IHC is chemical, specifically cannot withstand harsh retrieval methods and are
with formaldehyde. The time lapse between the death easily damaged, with subsequent loss of antigenicity.54
of the animal and the collection of tissues for IHC is
usually critical, and prompt transfer into fixative is Antigen retrieval
desired. Enzyme-rich tissues, such as intestine or
pancreas, autolyze rapidly. Protozoa and fungi may Antigen retrieval (AR) is intended to reverse the
be more resistant to autolysis than viruses. In general, detrimental effects of fixation. As mentioned, one
samples to be fixed should not be thicker than 0.5 cm, of the main effects of formalin fixation is
and the ratio of fixative to sample should be at least conformational changes in epitopes, but loss of
10 to 1. Factors influencing the quality of fixation electrostatic charges may occur during fixation and
include type of fixative, fixative pH and buffers, have an effect on antigen detection.16 The exact
fixative concentration, fixative osmolality, fixation mechanism by which AR works on formalin-fixed
time, fixation temperature, fixative additives, and the tissues is not clear. A variety of pathways may
use of additional post fixation procedures (e.g., contribute to its success, including the breaking of
decalcification). cross-linkages, the extraction of diffusible blocking
Coagulative fixation. Coagulative fixatives are proteins, the precipitation of proteins, the hydrolysis
organic and nonorganic solutions that coagulate of Schiff’s bases, calcium chelation, paraffin removal,
proteins and render them insoluble. The fixation and the rehydration of tissue, resulting in better
process removes and replaces free water, thereby penetration of antibody and increased accessibility to
Immunohistochemical techniques in veterinary diagnostic laboratories 397

Figure 3. Cellular location of antigens. Antigens can be located in one or multiple cell compartments. Knowledge of the expected
location of a particular antigen is essential to an adequate interpretation of the immunohistochemistry results. A, testis of a dog with
Sertoli cell tumor. Neoplastic cells have strong nuclear staining for GATA-3, which is the expected location for this antigen. Panels B–D
are examples of KIT distribution in mast cell tumors. KIT cellular distribution has been linked to mast cell tumor prognosis in dogs but
not in other species. B, skin from a horse with mast cell tumor. Neoplastic cells depict only membrane staining for KIT. C, skin from a
dog with mast cell tumor. The majority of cells show diffuse cytoplasmic staining for KIT. D, skin from a horse with mast cell tumor. A
mixed staining pattern including cytoplasmic membrane, cytoplasm, and paranuclear staining is observed in many cells.
Immunoperoxidase-3,39-diaminobenzidine (DAB). Bar 5 20 mm.
398 Ramos-Vara et al.

Figure 4. Effects of different antigen retrieval (AR) in the immunohistochemical reaction. Small intestine, dog. A, this section was
treated with proteinase K as AR resulting in no staining. B, the staining intensity in the muscle layer (m) is very strong but also the
background produced in the submucosa(s). A combined (proteinase K and heat-induced AR citrate) AR was used. C, heat-induced AR
with citrate was used and staining of the muscle layers (m) is intense. Background is minimal. Submucosa(s); muscularis mucosa
(arrowhead). All sections were stained by LSAB+ peroxidase-3,39-diaminobenzidine (DAB) for myosin smooth muscle. Bar 5 130 mm.
Figure 5. Labeled streptavidin–biotin peroxidase method. This 3-step method consists of primary antibody-binding tissue
antigens, a secondary biotinylated antibody recognizing the primary antibody, and avidin–peroxidase complexes that will bind the
secondary antibody via avidin–biotin bonds (reprinted from Ramos-Vara43 with permission from Allen Press).
Figure 6. Polymer-based technology. This is a 2-step method. The primary antibody will bind tissue antigens. The
immunoglobulin–peroxidase–polymer complex will bind via immunoglobulins to the primary antibody. This method does not involve
avidin and biotin molecules (reprinted from Ramos-Vara43 with permission from Allen Press).

antigen.37,62 Longer fixation times are thought to heat-induced AR, enzymatic digestion, and detergent-
decrease immunoreactivity and might require longer, induced AR (Fig. 4). For a practical approach to
harsher AR methods, increased antibody antigen retrieval, see the next section (‘‘Practical
concentrations or longer primary antibody approach to standardization of a new antibody’’).
incubation times to achieve a similar degree of
immunoreactivity. However, with the advent of Heat-induced antigen retrieval
heat-induced AR, retrieval of antigens in overfixed
tissues can still be achieved in many instances.45,48,51 The success of heat-induced AR is probably the
In a recent study using 30 antibodies targeting cellular result of reversal of formalin-induced chemical
antigens in different cell compartments and infectious modifications in protein structure. The source of
agents, only those to Canine parvovirus, Felid heat is not critical to the outcome, but a matter of
herpesvirus, and cytokeratin 7 had a significant convenience. Various types of equipment may be used
reduction in the signal after several weeks of fixation including a de-cloaker (commercial pressure cooker
(Webster J, Miller M, Ramos-Vara J: 2007, Proc with electronic controls for temperature and time),
AAVLD, p. 86). Background staining caused by harsh vegetable steamer, microwave oven, or pressure
AR methods is not uncommon, and sometimes cooker.7,20,32,41,63 Many veterinary diagnostic
precludes diagnostic evaluation of the staining. Three laboratories use a steamer and/or de-cloaker. The
main AR methods are currently used in FFPE tissues: advantage of a de-cloaker over other heating devices
Immunohistochemical techniques in veterinary diagnostic laboratories 399

is that the boiling temperature is not affected by the needed, as well as prolonged (e.g., overnight)
atmospheric pressure, which varies depending on the incubations with the primary antibody. Secondary
altitude. antibodies (linker reagents) and detection complexes
The relationship between temperature and expo- in commercially available detection kits do not need
sure time is inverse: the higher the temperature, the to be titrated. For practical purposes, when testing a
shorter the time needed to achieve beneficial results. new antibody, it is recommended to use 3 sets of 5
The pH of the retrieval solution is important.62 Some slides (15 slides total): one set without AR, one set
antibodies bind well regardless of retrieval solution with enzymatic digestion, and one set with heat-
pH, whereas others bind weakly at neutral pH, but induced AR. The first 4 slides in each set will have 2-
strongly at very low or high pH. Common buffers fold dilutions of the primary antibody and the fifth
used in heat-induced AR are citrate, Tris-HCl, and slide should be incubated with the negative reagent
EDTA (ethylenediamine tetra-acetic acid). A low pH control.44,54 A similar approach has been suggested by
buffer (acetate, pH 1.0–2.0) appears especially useful the College of American Pathologists.28
for nuclear antigens. The significance of the chemical Incubation conditions. The conditions for incu-
characteristics of the retrieval solution, particularly bation of the primary antibody depend on the
the buffer, is unclear. antibody characteristics (e.g., affinity), environmental
Enzymatic digestion. Enzymes are thought to factors (e.g., temperature), section characteristics, and
digest the tissue to some degree, allowing antibodies procedure. Incubation for most routine IHC protocols
to recognize antigenic sites. This digestion is is 30–90 min at room temperature. During incubation,
nonspecific and some antigens might be negatively the sections must be completely covered by an adequate
affected by this treatment.68 Commonly used enzymes volume of the antibody solution to prevent section
include trypsin, proteinase K, pepsin, pronase E, and drying and assure even exposure of the tissue to the
ficin. A combination of heat and protein digestion antibody solution. Shortened incubation times at 37uC,
may be necessary to demonstrate some antigens. longer incubation times at room temperature, or
Detergent-induced antigen retrieval. Detergents extended overnight incubation in a humidity chamber
solubilize membrane proteins by mimicking the lipid- at 4uC is occasionally utilized to achieve adequate
bilayer environment.54 They form mixed micelles that immunostaining.17 Additional tissue treatments (e.g.,
consist of lipids and detergents as well as detergent decalcification), length of fixation, and AR technique
micelles that contain proteins (also known as can affect incubation of the primary antibody.
surfactants), thereby decreasing the surface tension of Decalcification with weak acids does not seem to
water. They are usually incorporated into the dilution/ interfere significantly with immunostaining of most
rinse buffers. Examples of detergents include ionic antigens, provided the tissues were previously well fixed
detergents, bile acid salts, and nonionic and in formaldehyde. Strong acids may affect the detection
zwitterionic types (e.g., Triton R-X 100, BRIJR, of some antigens at various degrees.2,4,35 It is
Tween 20, saponin). recommended to use weak acidic decalcifying
solutions (e.g., formic acid) diluted in formalin.
Practical approach to standardization of a new antibody
a) Primary antibody characteristics.—There are
Dilutions of primary antibody. Antibody dilutions many characteristics of the antibody that affect
for one type of tissue may not work with different incubation time. The specificity of the antibody is
tissues for the same antigen (e.g., brain vs. lymphoid important in determining the length of incubation
tissue). Differences in processing techniques and other time to get sufficient binding. Lower-affinity anti-
variables may require antibody titration for each bodies require longer incubation times (and/or higher
tissue type. Often the manufacturing company has concentrations). Affinity rarely has to do with
data on antibody performance in FFPE tissue that whether the antibody is monoclonal or polyclonal.
can be used as a guideline for the starting dilution. Other antibody-dependent factors include immuno-
Typically 2- to 10-fold dilutions above and below the globulin isotype, manufacturer, clone, and lot differ-
manufacturer’s recommended dilution provide a good ences. Different lots of the same antibody may vary in
starting point. Depending on the antibody type concentration or other solution characteristics and
(monoclonal or polyclonal) and concentration, a thus require re-evaluation.
range of 1–5 mg/ml should be used for an initial b) Environmental factors.—Incubation tempera-
titration. The optimal working concentration for ture is a major determinant of incubation time. In
most antibodies is generally from below 1 mg/ml to general, as temperature increases, incubation time
3 mg/ml, depending on the detection system utilized. decreases. Some antibodies might be temperature-
If an antibody has not yet been tested, a systematic sensitive with decreased immunoreactivity at higher
approach using a wide range of dilutions might be temperatures. Sections with longer incubation times
400 Ramos-Vara et al.

may dry out if there is inadequate humidity in the methods are faster than most current avidin–biotin–
incubation chamber. Performing the incubation in based methods, do not produce the background
humidity chambers helps to alleviate this complication. generated by endogenous biotin, and have
c) Tissues from different animal species.—Species comparable or sometimes superior sensitivity to
of origin of the tissue can dramatically affect avidin–biotin methods. They are also more expensive.
reactivity. Interspecies variations in antibody reac- Additional detection systems. There are multiple
tions results from subtle changes in the amino-acid variations to the above-mentioned systems aimed at
sequence of a given antigen in a particular species. increasing the sensitivity of the reaction. Examples of
Even in the event of interspecies cross-reactivity, the highly sensitive methods are the tyramine–biotin
antibody affinity may be decreased. Prolonged method (very sensitive but can create high
incubation times, varied AR methods, and/or in- background sometimes) and immunorolling circle
creased antibody concentration may be needed to amplification, which is a nonavidin–biotin method.43,60
obtain optimal staining. Specificity of the antibody The most commonly used enzymes are peroxidase and
must be verified for every species tested. alkaline phosphatase.43,54,64
Preservation procedure. The type of tissue There are no rules of thumb in selecting a detection
preservation will likely affect the performance of the system. The choice of system will depend on several
primary antibody. Frozen sections typically require factors: 1) degree of expertise/experience of the
less incubation time than do FFPE tissue sections. technician; 2) type of antigens to be detected (some
Alcohol or acetone fixation has variable effects on the do not need very sensitive methods because they are
incubation characteristics of the primary antibody. abundant); 3) number of tests (antibodies) available
There are commercially available fixatives that (different antibodies may require different detection
reportedly preserve epitopes better for IHC systems); 4) species idiosyncrasies (e.g., amount of
techniques than the standard formaldehyde,43 but endogenous biotin in tissues); 5) budget; and 6) best
these fixatives can be more expensive and less suitable signal-to-noise ratio when combined with the AR
for routine microscopy. method used.
Choice of detection system. The sensitivity of IHC
tests depends mainly on the detection system used. As Test validation
a general rule, the more complex an IHC method, the Validation of a diagnostic test is, in general, the
more sensitive it is. One step or 2-step IHC procedures process of optimizing the test method (reagents and
are usually less sensitive than more complex, multistep protocols) and determining the performance charac-
procedures. Most detection systems currently used in teristics of the test (Fig. 7). Requirements for test
diagnostic laboratories are based on a color change validation are published for AAVLD accreditation
induced by an enzyme attached to the immuno- and provide useful general scientific definitions and
complexes bound to a tissue section, after reacting practical steps for classifying laboratory tests as
with its substrate and chromogen. The 2 most common ‘‘validated for use,’’ but are not specific for IHC
detection systems are avidin–biotin and polymer-based assays (Essential Requirements for an Accredited
nonavidin–biotin systems. Veterinary Medical Laboratory Version 4.1, Section
Avidin–biotin systems. These systems are based on 5.4.3, 2006. Published by AAVLD). For most IHC
the strong affinity of avidin for biotin. Avidin–biotin assays, this involves detecting any cross-reactivity of
systems are currently the most commonly used the selected antibody with unrelated antigens, and
detection systems. Biotin is usually attached to a cross-reactivity among different tissues and among
secondary antibody that binds a complex of avidin– different species. Moreover, validation examines the
enzyme to produce a colored reaction. These methods variables that affect the IHC reaction, such as fixation
are very sensitive (Fig. 5). Problems with endogenous time and storage of paraffin blocks or storage of
biotin background, particularly when using harsh unstained tissue sections. Last, but not least, valida-
(heat) AR methods or staining tissues especially rich tion of an IHC test may include comparison of results
in biotin (e.g., liver), may occur; blocking is usually among different laboratories using similar tech-
necessary but is also expensive. niques.9 When possible, validation compares the
Polymer-based nonavidin–biotin systems. These sensitivity of IHC detection to the gold standard
systems are usually 2-step procedures (can be method of detection for the antigen in question. It
increased to a 3-step procedure if more sensitivity is may also compare staining patterns and sensitivity
necessary; Fig. 6). The first step is the unlabeled with other antibodies targeting the same antigen.
primary antibody; the second step consists of a Validation is usually an ongoing process and gener-
polymer containing numerous secondary antibodies ally requires extensive testing. Although the AAVLD
as well as numerous molecules of enzyme.59,75 These IHC subcommittee believes that minimum standards
Immunohistochemical techniques in veterinary diagnostic laboratories 401

Figure 7. Validation of an immunohistochemical test. Factors to be included in the immunohistochemistry validation are antigen
cross-reactivity, species cross-reactivity, fixation, equipment used, storage of reagents, and test results.

for validation are needed, the exact validation belong to the same group, or produce similar lesions
procedure will depend on the selected antibody and in the organ of interest, be determined. For example,
the laboratory (in-house validation). As with stan- an antibody to Leishmania sp. should be tested against
dardization, one goal of validation is to produce the other protozoa (e.g., Trypanosoma, Eimeria, Isospora,
same result regardless of the IHC method used or the Cryptosporidium, Giardia, Neospora, Toxoplasma, Sar-
laboratory where the test is performed. The primary cocystis; Fig. 8) and morphologically similar fungi
aim among laboratories should be documentation of (e.g., Pneumocystis, Histoplasma, Sporothrix). For
their validation procedure(s) as it applies to their antibody-targeting viruses, it is necessary to test against
methods for the IHC detection of an antigen. other viruses within the same group that affect the
same tissue or produce similar lesions. For example, an
Cross-reactivity
antibody to Canine parvovirus should be tested in
Two main types of cross-reaction are considered: tissues infected with Canine coronavirus, Canine
antigen cross-reactivity and species cross-reactivity. It distemper virus, and Canid herpesvirus.
is assumed that testing conditions (e.g., fixation, Some lack of specificity does not necessarily
tissue processing, antigen retrieval, incubation times, preclude the use of an antibody provided it is
detection kits) are the same as those used during documented and can be interpreted. An example of
standardization of the IHC test. this would be the use of an antiserum raised against
Antigen cross-reactivity. The type of antigen (i.e., Measles virus nucleoprotein, which reacts with most
structure, amino-acid sequence) affects cross- known morbilliviruses, for the detection of Canine
reactivity. A literature search may reveal reports of distemper virus in dogs. Some mAb against infectious
antigen cross-reactivity. Lack of cross-reactivity in 1 agents will cross-react with cell organelles (Fig. 9).
species does not preclude this potential problem in b) Neoplastic diseases.—Developing IHC protocols
other species. for antibodies to specific cell types or cell components
a) Infectious diseases.—For antibody-targeting bac- requires one or more of the following comparisons:
teria, protozoa, and fungi, it is suggested that the
cross-reactivity of an antibody to other bacteria/ i. Comparison of immunoreactivity of primary
protozoa/fungi that are morphologically similar, tumors and their metastases.45,48
402 Ramos-Vara et al.

Figure 8. Antibody cross-reactivity. A, kidney from a horse infected with Bartonella henselae and stained with antibody to B.
henselae. Strong staining in areas containing microorganisms. B, the same tissue was stained with an antibody to Borrelia sp. The
staining is similar in intensity and anatomic location to that seen in panel A. A polymerase chain reaction of the affected tissue using
primers specific for Bartonella sp. and Borrelia sp. demonstrated genome amplification of only Bartonella. This is an example of the
cross-reactivity of some antibodies and the necessity for extensive validation studies to determine the degree of confidence of each
antibody. Glomerulus (g). Immunoperoxidase-3-amino-9-ethylcarbazole (AEC). Bar 5 35 mm.
Figure 9. Antibody cross-reactivity. In this case, an antibody recognizing rotavirus (arrowheads) in a porcine intestine also cross-
reacts with a supranuclear structure (interpreted as the Golgi apparatus) in most enterocytes (arrows). The authors have observed similar
nonspecific reaction with other monoclonal antibodies to Bovine viral diarrhea virus, Bovine coronavirus, and Bovine herpesvirus 1.
Immunoperoxidase-3,39-diaminobenzidine (DAB). Bar 5 17 mm.

ii. Determination of immunoreactivity in other species (Fig. 10). Detecting an antigen in a new
neoplasms that would be included in the differ- species may be difficult and laborious. Nevertheless, it
ential diagnosis (e.g., oral spindle cell sarco- is advisable to restandardize (optimize) a test for each
mas).46,48,50,53 species, including incubation times, concentration of
iii. Comparison of immunoreactivity between non- the primary antibody, and AR. A literature search or
neoplastic and neoplastic tissues (antigen expres- consultation with the antibody manufacturer and
sion may be lost or expressed de novo in the extensive testing of similar cases in a given species are
neoplastic phenotype).51 recommended to determine species cross-reactivity.
iv. Determination of differences in immunoreactiv- For diagnostic purposes, an IHC report should state
ity in tumors with variable cell phenotypes (e.g., the degree of confidence in the results based on the
well-differentiated and poorly differentiated) in laboratory’s or others’ experience (including pub-
the same tissue or different tissues (e.g., mela- lished material). The use of tissue arrays from
noma).45,48 different animal species permits rapid screening for
v. Determination of cross-reactivity of an antibody interspecies cross-reactivity.31,39,40,43 Other advantages
targeting a particular cell type in metastatic of the tissue microarray are savings in reagents and
tumors that can be present in the same organ reduction in results variability; disadvantages include
(e.g., hepatocellular tumors and tumors metasta- a) selection of the sample is critical because of its
sizing to the liver).49 small size, and b) highly qualified personnel are
vi. Determination of differences in reactivity be- needed to prepare the array.
tween native and mutated antigens (e.g., p 53). Effects of fixation, postfixation treatments, and equipment
vii. Determination of prevalence of immunoreactiv- on immunoreactivity
ity of various antibodies in a variety of tumors or
tissues to determine the relative utility of a Different fixatives have different effects on immuno-
particular antibody for supporting the diagnosis reactivity. For each fixative, full standardization of a
of a cell type based on the IHC reaction.51,53 test must be done, including incubation conditions,
dilution of the primary antibody, and AR method
Species cross-reactivity. This problem is unique to (see ‘‘Standardization’’ section). Standardization is
veterinary medicine. Although some antigens can be usually done on tissues with known fixation time
detected under similar conditions in different species, (e.g., 1–2 days).
it should be assumed that antigen detection will vary Fixation kinetics studies are recommended for each
among species.52 Identical antibody clones that target antigen or epitope. A positive tissue control should be
the same antigen can differ in reactivity among fixed for different durations (e.g., 1, 2, 4, 7, 10, 14, 21,
Immunohistochemical techniques in veterinary diagnostic laboratories 403

20 days) and tested under identical conditions to seems semantic but underscores not only the need to
determine the effects of fixation time on reactivity interpret subjectively a colored reaction, but to do so in
(i.e., intensity and number of cells or organisms the context of the disease.61
detected; Fig. 11).45,48
Automated stainers are designed to duplicate Controls
manual staining procedures and can be used to ensure Positive tissue control
uniform application of all steps of the process. Thus,
the use of automated equipment offers a uniform and Positive tissue control is defined as tissue that is
standardized microenvironment for testing, which will, known to contain the antigen of interest detected by
in turn, result in intralaboratory run-to-run consist- identical IHC methods to those used in diagnostic
ency.64,77 When results among different laboratories cases. Fresh-fixed surgical specimens (or biopsy
differ considerably, these technical differences should tissue) are preferred over necropsy material whenever
be considered as a possible cause. possible, since autolysis may affect staining results.
Positive tissue controls must be fixed and stained in
Storage of paraffin blocks and tissue sections the same way as the diagnostic case tissue for every
It is the authors’ and others’ opinion that paraffin antibody and procedure used.55,64 The presence of the
blocks remain stable for years in terms of antigenicity. antigen in the tissue control should be confirmed by
However, only controlled studies on the effect of another method (e.g., PCR, virus isolation). For most
nucleic acid detection on archival paraffin blocks laboratories, control tissues are generally obtained
have been published.25 Any deleterious effects of ‘‘in-house’’ because variables such as fixation and
prolonged storage on tissue antigenicity in paraffin processing are the same as those used with diagnostic
blocks, if they happen, may differ among antigens. case material. Control tissues obtained from another
For practical purposes, consider this factor if laboratory or from a commercial source may have
inconsistent IHC results occur when using old been fixed/processed in a different way than the test
paraffin blocks. tissue. However, the use of in-house standards does
Storage of unstained paraffin control tissue sec- not ensure reproducibility of a particular IHC assay
tions increases efficiency but may adversely affect between different laboratories.
immunoreactivity (tissue section aging), depending on Multi-tissue blocks and cell lines. Various methods
the antigen of interest.24 Photo-oxidation of tissue for the simultaneous study of multiple FFPE tissues
sections is involved in loss of antigenicity.13 Tissue in a single histologic section have been reported. The
section ageing is a rather common problem with ‘‘sausage technique’’ has been used heavily in the
nuclear antigens (e.g., Ki67, estrogen receptor, past.10 In this method, long thin strips of fixed tissues
p53).11,19,25,30,34,36,38,42,54,70,72 When there is decreased are drawn into a tube of unfixed small intestine. The
intensity or loss of reaction in stored control tissue entire sample is then fixed, resulting in a tight column
sections, repeat the test with another stored control that can be cut into blocks and embedded. Paraffin-
tissue slide and a freshly cut control tissue section. If embedded tissue microarrays have been proposed as a
the change is limited to stored sections, discard any reference control standard for human IHC laborato-
remaining unstained control slides. ries, primarily for use in tumor diagnosis.29,39,40,43 It is
recommended that validation studies be carried out
What constitutes a positive/significant result? on multi-tissue control blocks containing both
There is no single answer to this question. Immu- known-positive and known-negative normal and
nohistochemical assays detect analytes (infectious tumor tissues.64 In veterinary medicine, species-
agent or tumor marker antigens) in tissues but do specific tissue microarrays are not commercially
not necessarily diagnose disease, which may require available. Species-specific tumor cell lines are being
interpretation of IHC results in correlation with other used for IHC controls (e.g., IHC for HER-2/neu).55,57
clinical or laboratory findings. For an infectious agent, This approach provides an identical tissue control
detecting one organism indicates infection, but it is among different laboratories and is an excellent way
another matter to prove that the agent caused disease. to compare results among them.
For neoplasms, this answer is even more difficult. In Reference control tissues for infectious diseases. The
the literature, the percentage of positive cells required presence or absence of a particular infectious agent in a
to confirm tissue origin of a tumor varies considerably, tissue should be assessed by previously published
and often this striking variation is reported for IHC reactivity patterns (if available) plus another
detection of the same antigen by different authors. non-IHC diagnostic method.8 For example, Listeria
Some pathologists prefer the words ‘‘detected’’ or ‘‘not monocytogenes–control tissue could be assessed
detected’’ rather than ‘‘positive’’ or ‘‘negative.’’ This by IHC (immunoreactivity of extracellular or
404 Ramos-Vara et al.

Figure 10. Species reactivity differences. Eyelid, horse. This tissue was incubated with two different antibodies to vimentin, a
mouse monoclonal antibody (A) and a rabbit monoclonal antibody (B). Reactivity is only observed in the tissue incubated with the
mouse monoclonal antibody. There is spurious staining in the mucosal epithelium in panel B (asterisk). The rabbit monoclonal antibody
to vimentin reacts as expected in other animal species (e.g., dog, cat). Immunoperoxidase-3,39-diaminobenzidine (DAB). Bar 5 65 mm.
Figure 11. Effects of prolonged fixation. A, tissue was fixed for 24 hr. B, tissue was fixed for 11 weeks. Bartonella organisms
(asterisks). Note that there is no staining in tissue fixed for several weeks. Sensitivity to fixation is variable for each antibody; thus, the
need to evaluate fixation kinetics in any new immunohistochemical procedure, if possible. Immunoperoxidase-3,39-diaminobenzidine
(DAB) for Bartonella. Bar 5 33 mm.
Immunohistochemical techniques in veterinary diagnostic laboratories 405

phagocytized bacilli) and by bacterial culture. performed on the tissues/organ systems of the same
Similarly, Bovine viral diarrhea virus (BVDV)–control animal should be used to rule out the presence of the
tissue could be assessed by IHC (immunoreactivity in antigen of interest. Both positive and negative in-
cytoplasm of infected cells) and by virus isolation or house tissue controls should be used in each test and
PCR. In other instances, the presence of characteristic should be processed in the same manner as the case
lesions for a particular disease (e.g., intranuclear material. Similarly, when using multitissue blocks for
inclusions observed with hematoxylin and eosin [HE] antibody validation studies or as tissue controls, the
staining) can be used as a standard reference control. specimens must be fixed and processed in the same
Immunohistochemical protocols for infectious dis- manner as the case material.64 When dealing with
eases should, whenever possible, also be species- cellular antigens (e.g., cytokeratin, vimentin), positive
matched with test specimens; alternatively, tissues control tissue should have areas expressing variable
from other species affected with the same infectious amounts of the specific antigen. Weakly stained areas
agent can be used (e.g., BVDV identified in diagnostic can be used to detect subtle changes in antibody
case material from cervids using a bovine control sensitivity.64 In practice, only 1 tissue control is used
tissue). Nonspecific binding of primary antibody because of the common presence of both positive and
(species-specific molecular mimicry) or secondary negative cells within the control.
(linker) reagents may occur among different species.
A good example is the use of goat polyclonal Internal positive tissue controls
antiserum specific for Neospora caninum on goat Internal positive tissue controls are present in
tissue. The anti-goat IgG secondary reagent used in diagnostic case tissues. An example is the detection
the IHC assay can bind excessively to endogenous of smooth muscle markers or vimentin in normal
goat IgG in the tissue resulting in such severe blood vessels. The presence of positive staining in
background ‘‘noise’’ that visualization of individual these areas indicates appropriate immunoreactivity.
immunoreactive tachyzoites is impossible. With this type of control, there is no fixation variable
Reference controls for neoplastic diseases. Many between the control tissue and the diagnostic case
times, the only way to verify the presence of an tissue.14 Although some vimentin antibodies have
antigen is by morphology, which is not always an been used to demonstrate overfixation effects due to
accurate predictor for the presence or absence of a their sensitivity to fixation,55 this type of test may not
particular antigen. Furthermore, a negative result for be relevant with the current AR methods.
tumor markers does not necessarily rule out a
particular neoplasm because the cell marker of Reagent (antibody) controls
interest may not be expressed in a very poorly Negative reagent controls are used to confirm the
differentiated cell population. Ideally, controls for specificity of the test and to assess the degree of
tumor cell markers should consist of the neoplastic nonspecific background staining present by omitting
tissue of interest, which expresses different intensities the primary antibody. Commonly, the primary
of reactivity.64 Control tissues for IHC tumor markers antibody is replaced by 1 of the following methods:
should also be species-matched with test specimens 1) antibody diluent, 2) same species nonimmune
because the detection of an antigen in a particular immunoglobulin of the same dilution and immuno-
tumor and animal species does not guarantee similar globulin concentration, 3) an irrelevant antibody, or
results in a different animal species. 4) buffer.55,64 These methods will assess the degree of
cross-reactivity of the primary antibody, and the
Negative tissue control degree of nonspecific binding by the labeling (sec-
Negative tissue control is defined as tissue that is ondary) antibody and detection system; only methods
known not to contain the antigen of interest.64 At 2 and 3 approach the true significance of negative
least 1 ancillary test (e.g., PCR, virus isolation) controls. There are also commercially available ready-

Figure 12. Nonspecific staining. Knowledge of the expected antigen distribution of the reaction within a cell or tissue is mandatory
before using a test for diagnostic purposes. In this case, an antibody to CD79a stained only the nucleus of plasmacytoma cells. This
reaction is not considered specific based on the current knowledge of the location of this antigen (cytoplasmic and cytoplasmic
membrane). Bar 5 60 mm. The lower inset is a detail of the aberrant staining; the upper inset depicts a typical cytoplasmic staining of
CD79a in a plasmacytoma. Immunoperoxidase-3,39-diaminobenzidine (DAB). Both inset bars 5 5 mm.
Figure 13. Equipment maintenance. One of the possible causes of unusual staining in immunohistochemistry is inadequate
maintenance of equipment. The heat unit (arrows) of the steamer needs to be free of salts that will progressively deposit when using tap
water. A, clean unit. B, unit with severe salt deposition. The heat unit in panel B could not reach the expected temperature.
406 Ramos-Vara et al.

to-use universal negative reagents for rabbit and enzyme inhibition by NaN3, use 0.01%
mouse primary antibodies. merthiolate in conjugates.26,66 Do not use
If the diagnostic workup requires a panel of preservatives in buffers or antibody diluents.
antibodies each of the same isotype and similar b. Close inspection of buffers and other re-
concentration and derived from the same species, the agents for signs of microbial contamination
panel of antibodies itself may serve as a set of or precipitation of salts is necessary each time
irrelevant reagent controls; thus, the need for multiple the reagent is used.
negative controls is eliminated. If this method is
4. Proper labeling of stored reagents is essential.
utilized, separate controls should still be run for each
Each reagent label should include the name of
different type of protocol (e.g., for each protocol with
reagent, date made or opened, expiration date,
a difference in the AR method or difference in the
and name or initials of individual who made or
detection system).64
opened the reagent.
Storage and handling of reagents 5. Stock solutions often have a longer storage life
compared to working solutions. Working solu-
The shelf life of many reagents is directly linked to tions can be prepared daily for immediate use or
appropriate storage. The shelf life of many primary stored for short term.
antibodies beyond that indicated by the manufacturer 6. Substrates (or chromogens) should be freshly
can be significantly extended with proper handling and prepared for each run or as directed by the
storage.6,67 However, use of a reagent beyond the manufacturer.
manufacturer’s expiration date is not covered by its 7. Storage at 270uC does not provide significantly
warranty and has to be properly documented. General longer shelf life than storage at 220uC. Storage of
considerations for storage of reagents include: reagents in frost-free freezers is not recommended
1. Storage containers must be made of nonreactive because of their programmed fluctuations in
material that does not alter the reagent by temperature that contribute to increased polymer-
adsorption or polymerization of components ization and denaturation of many proteins.
within the reagent or addition of components to 8. Routine temperature checks of all storage areas
the reagent. and appropriate relevant documentation are
recommended.
a. Plastics: Polypropylene and polycarbonate
are nonreactive and are routinely used for
Quality assurance/quality control and interpretation of
storage of primary antibodies at 220uC or
IHC results
270uC. Containers that can be tightly sealed
are necessary to prevent desiccation.18 Quality reflects on the procedural and technical
b. Borosilicate glass: This material is used when aspects of the IHC test and is aimed to reduce and
storing reactive reagents. When storing di- correct deficiencies in an analytical process. Each step
luted antibody preparations, addition of of an IHC test is defined by quality control (QC)/
0.1–1.0% bovine serum albumin (BSA) may quality assurance (QA) standards that have been
be necessary to decrease polymerization and previously addressed. Quality control issues, as they
adsorption in any container used.26 pertain to daily records and the standardization of
new primary and secondary antibodies and controls,
2. Primary antibodies should be stored frozen as
are discussed here.
concentrates in appropriately sized aliquots in
keeping with manufacturer’s recommendations. Daily quality assurance/quality control
Freezing minimizes denaturation of proteins and
Each laboratory should establish standard opera-
reduces excessive mechanical action or contact
tion procedures (SOPs) for their routine histology
with air.26 Once thawed and diluted, the antibody
laboratory. This would include daily, weekly, and
preparation should be stored at 2–8uC. Repeated
monthly schedules for cleaning, maintenance, and
freezing and thawing of the concentrated anti-
monitoring logs, along with a daily check of
body can result in significant loss of reactivity.
equipment such as oven temperature for drying slides,
3. The majority of buffers are stored at 2–8uC to
temperature of water baths, pH meter, etc. Buffers
prevent microbial growth.
should be made using distilled deionized water
a. Sodium azide (NaN3) is often used in (ddH2O) and their pH checked and adjusted, if
commercial concentrated primary antibody necessary, before use. When enzymes are used for
preparations as a preservative. For in-house AR, they should be prepared shortly before use.
antibodies, use 0.02% NaN3. To avoid There are also ready-to-use commercial enzyme
Immunohistochemical techniques in veterinary diagnostic laboratories 407

solutions for AR. Note the storage conditions and the pathologist and a written evaluation of the quality
expiration dates for any reagent. of the previous interpretation should be provided.
A worksheet should list the stains requested and Written comments on the technical aspects and the
number of slides to be stained. The slides should be overall quality of the slides should be made.
labeled accordingly and double-checked against the
IHC request forms submitted by the pathologist. External quality assurance/quality control
Fresh working solutions of primary antibody The goal for interlaboratory comparisons is to
should be prepared according to their storage document variations in reactivity (e.g., positive vs.
requirements. The expiration dates of the antibody negative, intensity, number of positive cells) among
and the working dilution should be indicated on the different laboratories and to set minimum QA/QC
tube. Xylene/xylene substitute and graded alcohols standards to create optimal protocols that could be
used for deparaffinization, rehydration, and dehydra- shared by all laboratories.74 Interlaboratory stan-
tion should be changed before they lose significant dardization is difficult. Even in human medicine, only
strength. It is recommended to use AR solutions only a handful of IHC tests (e.g., Hercep test) have been
once; however, each laboratory should establish their validated in such a way that different laboratories
own protocol for reusing AR solutions. could perform tests with consistent results, and not
When using an automatic stainer, a ‘‘Before without controversy.12,27,58 Veterinary diagnosticians
Staining Checklist’’ should be observed. are still a long way from achieving interlaboratory
The following are some important items to include standardization. As a previous analysis pointed out,
on the list: ‘‘Whether we like it or not, the practice of anatomic
pathology is to some extent subjective, although we
N Check to ensure there is room for waste; empty
all strive for as much objectivity and reproducibility
the waste containers if necessary.
as possible in our daily work.’’23
N Check quantities of reagents for the run: ddH2O,
There are 2 main approaches to interlaboratory
buffers, IHC kit reagents, and chromogen.
standardization:
N Check to ensure correct programming is being
used for the run. 1. Technical equivalency testing of an IHC test.
N Check slide labels as the stainer is loaded. Unstained slides are distributed among different
N Check the quantity and placement of the IHC laboratories for performance of the requested
reagents. immunostains; results are compared among dif-
N Remove all caps from reagent tubes. ferent laboratories or with those of the reference
laboratory. This test determines the quality and
The maintenance schedule for the automated stainer
interlaboratory consistency of the IHC testing.
should be followed and documented.
2. Diagnostic equivalency testing using IHC testing as
Primary antibodies. The date received, date
an adjunct. In this case, both the pathologist’s
aliquoted, date of dilution, and expiration date must
proficiency and the quality of IHC testing are
be written on the appropriate containers. New lots of
evaluated. A brief history and description of the
antibody should be tested and titrated before use. It is
tissue examined is included with tissue sections. The
best to prepare only the quantity of reagents required
pathologist must make a diagnosis based on routine
in a given run. Aliquots should be made of stock
(e.g., HE) staining and support it with appropriate
primary antibody in volumes convenient for
IHC testing. Results could be reviewed by an
preparation of working dilutions.
established committee that would address possible
Secondary reagents. Most reagents are commer-
differences among participating laboratories.73
cially prepared and standardized by the manufacturer.
Reagents should be stored per manufacturer’s recom- The current working proposal of the Pathology
mendations. Batch numbers should be recorded. Keep Committee for the AAVLD Program for Interlabora-
in mind that guarantee of a reagent only applies when tory Comparison of Infectious Agent Immunohisto-
the product is used as specified by the manufacturer. chemical Assays recommends having a reference
Control tissues and stains. See ‘‘Controls’’ section. laboratory (proposed to be the National Veterinary
Laboratory Services, Pathobiology Section, Ames,
Internal quality assurance/quality control IA) send participating laboratories a set of unstained
A bi-annual internal QA/QC check for the quality FFPE sections for immunostaining and interpreta-
of IHC slide interpretation is recommended. In large tion. Results should be qualitative (detected/positive,
diagnostic laboratories, randomly selected slides not detected/negative) and not quantitative. Partici-
should be reviewed by other pathologists on a pating laboratories would return the slides and
rotational basis. The slides should be interpreted by interpretations to the reference laboratory for evalu-
408 Ramos-Vara et al.

ation and feedback. For cellular antigens, a similar 2. Review assay logs/datasheets/checklists for ap-
approach may be considered in the future. propriate incubation times and temperatures.
3. Check protocol for appropriate pretreatment or
Immunohistochemical report and interpretation AR.
If not included in the standard report format, the 4. Make sure the linking antibody is compatible
IHC report should contain demographic information with the primary antibody.
pertinent to the case, the tissue that was tested, and 5. Check for out-of-date reagents or antibodies and
the antibody used, or the antigen in question should ensure that they have been properly stored
be listed. Other details of the IHC test should be filed according to package inserts.
in the laboratory. Results for infectious antigens have 6. Ensure that the primary antibody was properly
to be reported as ‘‘detected’’ or ‘‘not detected.’’ In diluted.
addition, the interpretation for infectious disease 7. If a reagent was stored frozen, determine that it
IHC/immunocytochemistry (ICC) tests should in- was not repeatedly frozen and thawed.
clude a disclaimer that any lack of detectable antigen 8. Check deparaffinization protocols and reagent
may be a result of the absence of the antigen in this quality (e.g., not overused).
particular section and/or caused by technical aspects, 9. Ensure that the substrate-chromogen was freshly
such as prolonged fixation. In the case of tumor and properly prepared.
markers, it may be important to include a description 10. Determine that the slides did not dry out at any
of the cellular location (e.g., cytoplasmic, membrane, time during the procedure.
nuclear) and intensity of staining and percentage of 11. Check the pH of the buffer solutions.
positive cells, along with an interpretation of the 12. Other less common but possible problems:
significance of the test results (Fig. 12). When the
antigen location is atypical, the report should a. Sodium azide in the wash buffers may inhibit
mention this. Personnel who read IHC slides should peroxidase reactions.
be familiar with the antibodies used and their b. Certain commercial phosphate buffer solutions
specificity and sensitivity. The authors highly recom- may inhibit alkaline phosphatase activity.
mend having a trained veterinary pathologist for QA/ c. Use of inappropriate counterstain or dehydra-
QC purposes review daily IHC staining. tion steps with alcohol-soluble chromogens.
d. Exposure of tissues to temperatures of .60uC
Troubleshooting guide during embedding or drying.
Weak or no staining of positive control and e. Leaving too much buffer on slides after
weak or no staining of test slides washes. This dilutes reagents.
Consider the following: f. Excessive counterstaining interfering with
interpretation.
1. Problem: all slides from some primary antibodies g. Prolonged storage of tissue sections.
in the run are affected. Action: check for
inadequacy of the primary antibody, method 13. Try a new positive control tissue.
incompatibility, primary and link antibody in-
compatibility, or inadequate AR (Fig. 13). Inadequate or no staining of test slide and
2. Problem: the whole run is affected. Action: check adequate staining of positive control slide
assay log and adequacy of reagent volumes and
sequence of reagent delivery to the slide. Deter- 1. Problem: this is a true negative result; the antigen
mine if reagents were delivered to all slides (e.g., in question is not present in the test tissue.
buffer, chromogen). Action: none.
3. Problem: if it is hit-and-miss throughout the run, 2. Problem: the antigen is present in the test tissue,
consider technical problems or problems with the but in a concentration below the method’s
tissues. Action: check for inadequate sequence of detection limits. Action: consider using an ampli-
reagents, unbalanced autostainer, and inadequate fication procedure, or increasing the primary
drop zone. antibody concentration, incubation time, or
temperature or a combination thereof.
Checklist assuming that the positive control tissue
3. Problem: the test tissue is over- or underfixed.
has been proven:
Action: modify AR protocol.
1. Review assay logs/datasheets/checklists to be sure 4. Problem: the test tissue is from a different species
that no step was omitted and that all steps were than the control tissue and has different reactiv-
done in correct sequence. ity with the primary antibody. Action: validate
Immunohistochemical techniques in veterinary diagnostic laboratories 409

the IHC test with same species control and test 6. Inappropriate AR used. Action: re-evaluate AR
tissues. conditions.
5. The following are other potential but uncommon 7. Incubation temperature too high. Action: reduce
problems: temperature.
Blocking problems.
a. Excessively high (.60uC) embedding or
drying temperature was applied to the test 1. Endogenous enzyme activity not suppressed.
slide only (assuming the positive control Action: increase concentration of blocking agent.
tissue and the test tissue are on different 2. Inadequate protein blocking. Action: use a
slides). Action: check oven temperature. different blocking agent.
b. Prozone effect occurred due to high concen- 3. Inadequate blocking of endogenous avidin-bind-
tration of the primary antibody in the test ing activity. Action: use an avidin–biotin blocking
tissue. Action: retitrate the primary antibody step or use a nonavidin–biotin detection method
for the test slide. (Fig. 14).
c. A technical error (e.g., inadequate determi- 4. Inadequate blocking of endogenous biotin. Ac-
nation of the drop zone by the autostainer; tion: use an avidin-biotin blocking step or a
unbalanced platform of autostainer or slide nonavidin–biotin detection method.
rack(s); insufficient amount of reagents) has 5. Blocking serum from improper species. Action:
occurred. This may occur on the test slide but use blocking serum from same species as the link
not the positive control slide (assuming the (secondary) antibody.
positive control tissue and the test tissue are
Primary antibody problems.
on different slides) or the test tissue but not
the positive control tissue (assuming the test 1. Primary antibody too concentrated. Action:
tissue and positive control tissue are on the retitrate primary antibody (Fig. 15).
same slide). Action: check autostainer balance 2. Primary antibody incubation time too long.
status and dropping zone, respectively. Action: reduce incubation time.
3. Primary antibody is from a similar or identical
No staining of positive control slide and species as the test tissue (e.g., mouse on mouse
adequate staining of test slide [MOM], rat on mouse). Action: use specific
protocols (i.e., MOM or similar kits) or addi-
1. Problem: technical error in the staining or tional blocking steps.
handling of the positive control slide. Action: the 4. Inadequate buffer washes (inappropriate buffer
log or checklist should be reviewed; if everything ion concentration). Action: modify ionic strength
is in order, the assay should be repeated. For of the buffer solution.
control blocks for infectious diseases, the antigen
to be tested may not be present through the entire Secondary antibody problems.
block. 1. Secondary antibody and label concentration too
2. Problem: tissue section aging. Lack of staining in high.
the tissue section control might be a result of tissue 2. Secondary antibody and label incubation time
section aging. Action: compare staining of stored too long.
tissue sections with a fresh cut control section. 3. Buffer washes insufficient.
4. Secondary antibody recognizes endogenous (tis-
Excessive background staining sue) immunoglobulins (Fig. 16).
5. Microbial contamination of primary antibody
Prestaining problems.
solution. Action: reformulate from fresh stock.
1. Inadequate fixation, necrosis, and autolysis Add antimicrobial preservatives. Respect proper
2. Tissue sections allowed to dry out. Action: reduce storage conditions for antibody solutions.
incubation time; incubate in a humidified chamber.
Chromogen and counterstain problems.
3. Sections not completely deparaffinized. Action:
use fresh dewaxing solutions. 1. Chromogen concentration too high. Action:
4. Slide adhesive inappropriate or too thick. Action: reduce concentration of chromogen.
use adhesives specific for IHC or positive- 2. Chromogen allowed to react too long. Action:
charged slides. reduce incubation time with chromogen.
5. Tissue section too thick. Action: prepare thinner 3. Buffer washes insufficient. Action: prolong buffer
sections. washes.
410 Ramos-Vara et al.

Figure 14. Background caused by endogenous avidin–biotin activity (EABA). Kidney, dog. A, strong background staining
(EABA) on a section incubated with negative reagent control and an avidin–biotin detection system. B, a consecutive tissue section
incubated with a reagent negative control and EnVision+, a nonavidin–biotin detection method. There is no background staining. C,
staining for cytokeratins in renal epithelium. There is no background staining. EnVision+ peroxidase-3,39-diaminobenzidine (DAB). Bar
5 60 mm.
Figure 15. Background staining caused by antibody concentration. Mammary gland, dog. A, the primary antibody to p63 is too
concentrated and immunohistochemical reaction is present not only in the nucleus, but also in the cytoplasm of epithelial and stromal
cells. B, the antibody concentration was optimized and only nuclei display staining (the nucleus is the typical location of this antigen).
Immunoperoxidase-3,39-diaminobenzidine (DAB) for p63. Bar 5 60 mm.
Figure 16. Background staining caused by secondary (link) antibody. This is a common problem when using secondary antibodies
recognizing antibodies from the same species as the tissue examined. In this case, 2 detection systems (LSAB+ for A and B) and
EnVision+ (for C and D) were used. Goat tissue stained for cytokeratins using LSAB+ detection system (the secondary antibody
recognizes endogenous goat immunoglobulins) has a strong nonspecific staining in both the positive (A) and negative (B) reagent
sections. Specific staining for cytokeratins in epithelial cells (arrowheads); nonspecific staining in the stroma (asterisk). Note that the
negative reagent control section (B) does not have staining in epithelial cells. C, when similar tissues were stained with the same antibody
using EnVision+ as detection system (it does not recognize goat immunoglobulins), only specific staining of epithelial cells was observed.
D, as expected, the negative reagent control does not show staining. Immunoperoxidase-3,39-diaminobenzidine (DAB) for cytokeratins.
Bar 5 60 mm.
Immunohistochemical techniques in veterinary diagnostic laboratories 411

4. Counterstain obscures the IHC reaction. Action: virginianus) and mule deer (Odocoileus hemionus). J Vet Diagn
use a different counterstain that does not Invest 18:147–155.
10. Battifora H: 1986, The multi-tumor (sausage) tissue block.
interfere with IHC staining. Novel method for immunohistochemical antibody testing.
An excellent review of troubleshooting is available by Lab Invest 55:244–248.
11. Bertheau P, Cazals-Hatem D, Meignin V, et al.: 1998,
Atwood in Immunohistochemical Staining Methods.5 Variability of immunohistochemical reactivity on stored
paraffin slides. J Clin Pathol 51:370–374.
Conclusion 12. Bilous M, Dowsett M, Hanna W, et al.: 2003, Current
Immunohistochemistry is a well-established ancil- perspectives on HER2 testing: a review of national testing
guidelines. Mod Pathol 16:173–182.
lary technique to facilitate the diagnosis of infectious
13. Blind C, Koepenik A, Pacyna-Gengelbach M, et al.: 2008,
and neoplastic processes in animals. This article has Antigenicity testing by immunohistochemistry after tissue
reviewed the main factors involved in the IHC test, oxidation. J Clin Pathol 61:79–83.
from the preservation of samples and preparatory 14. Boenisch T: 2001, Formalin-fixed and heat-retrieved tissue
steps, followed by the immunohistochemical reaction antigens: a comparison of their immunoreactivity in experi-
and visualization of the reaction, to the interpretation mental antibody diluents. Appl Immunohistochem Mol
Morphol 9:176–179.
and generation of an IHC report. Each step of the 15. Boenisch T: 2005, Effect of heat-induced antigen retrieval
IHC test may require troubleshooting and a guide for following inconsistent formalin fixation. Appl Immunohisto-
this is also included. One goal of this article is to chem Mol Morphol 13:283–286.
suggest guidelines to help veterinary laboratories 16. Boenisch T: 2006, Heat-induced antigen retrieval: what are we
establish standardization and validation procedures retrieving? J Histochem Cytochem 54:961–964.
17. Boenisch T: 2006, Basic immunohistochemistry. In: Immuno-
in diagnostic IHC, thus providing a method of quality
histochemical staining methods, ed. Key M, 4th ed., pp. 15–
control and quality assurance in a very subjective 25. Dako Corporation, Carpinteria, CA.
discipline. 18. Carson FL: 1997, Histotechnology: a self-instructional text,
2nd ed. ASCP Press, Chicago, IL.
Acknowledgements 19. Di Vito KA, Charette LA, Rimm DL, Camp RL: 2004, Long-
term preservation of antigenicity on tissue microarrays. Lab
The authors are members of the AAVLD Subcommittee
Invest 84:1071–1078.
on Standardization of Immunohistochemistry. 20. Elias JM: 2001, Antigen restoration. The ‘‘hot’’ revolution in
immunohistochemistry. J Histotechnol 24:193–198.
References 21. Eltoum I, Fredenburgh J, Grizzle WF: 2001, Advanced
1. Anonymous: 1999, Quality assurance for immunocytochem- concepts in fixation: 1. Effects of fixation on immunohisto-
istry; approved guideline. NCCLS document MM4-A, chemistry, reversibility of fixation and recovery of proteins,
Wayne, PA. nucleic acids, and other molecules from fixed and processed
2. Arber JM, Arber DA, Jenkins KA, et al.: 1996, Effect of tissues. 2. Developmental methods of fixation. J Histotechnol
decalcification and fixation in paraffin-section immunohisto- 24:201–210.
chemistry. Appl Immunohistochem 4:241–248. 22. Eltoum I, Fredenburgh J, Myers RB, et al.: 2001, Introduction
3. Arnold MM, Srivastava S, Fredenburgh J, et al.: 1996, Effects to the theory and practice of fixation of tissues. J Histotechnol
of fixation and tissue processing on immunohistochemical 24:173–190.
demonstration of specific antigens. Biotech Histochem 23. Fletcher CD, Fletcher JA: 2002, Testing for KIT (CD117) in
71:224–230. gastrointestinal stromal tumors: another Hercep test? Am J
4. Athanasou NA, Quinn J, Heryet A, et al.: 1987, Effect of Clin Pathol 118:163–164.
decalcification agents on immunoreactivity of cellular anti- 24. Grabau DA, Nielsen O, Hansen S, et al.: 1998, Influence of
gens. J Clin Pathol 40:874–878. storage temperature and high-temperature antigen retrieval
5. Atwood KN: 2006, Troubleshooting. In: Immunohistochem- buffers on results of immunohistochemical staining in sections
ical staining methods, ed. Key M, 4th ed., pp. 131–155. stored for long periods. Appl Immunohistochem 6:209–213.
DAKO Corporation, Carpinteria, CA. 25. Guerrero RB, Batts KP, Brandhagen DJ, et al., Effects of
6. Balaton AJ, Drachenberg CB, Rucker C, et al.: 1999, formalin fixation and prolonged block storage on detection of
Satisfactory performance of primary antibodies beyond hepatitis C virus RNA in liver tissue. Diagn Mol Pathol,
manufacturer’s recommended expiration dates. Appl Immu- 6:277–281.
nohistochem Mol Morphol 7:221–225. 26. Harlow E, Lane D, ed.: 1988, Storing and purifying antibodies.
7. Bankfalvi A, Navabi H, Bier B, et al.: 1994, Wet autoclave In: Using antibodies: a laboratory manual, pp. 283–318. Cold
pretreatment for antigen retrieval in diagnostic immunohisto- Spring Harbor Laboratory Press, New York, NY.
chemistry. J Pathol 174:223–228. 27. Hornick JL, Fletcher CD: 2002, Immunohistochemical
8. Baszler TV, Evermann JF, Kaylor PS, et al.: 1995, Diagnosis staining for KIT (CD 117) in soft tissue sarcomas is very
of naturally occurring bovine viral diarrhea virus infections in limited in distribution. Am J Clin Pathol 117:188–193.
ruminants using monoclonal antibody-based immunohisto- 28. Hsi ED: 2001, A practical approach for evaluating new
chemistry. Vet Pathol 32:609–618. antibodies in the clinical immunohistochemistry laboratory.
9. Baszler TV, Kiupel M, Williams ES, et al.: 2006, Comparison Arch Pathol Lab Med 125:289–294.
of two automated immunohistochemical procedures for the 29. Hsu FD, Nielsen TO, Alkushi A, et al.: 2002, Tissue
diagnosis of scrapie in domestic sheep and chronic wasting microarrays are an effective quality assurance tool for
disease in North American white-tailed deer (Odocoileus diagnostic immunohistochemistry. Mod Pathol 15:1374–1380.
412 Ramos-Vara et al.

30. Jacobs TW, Prioleau JE, Stillman IE, et al.: 1996, Loss of monoclonal antibody hepatocyte paraffin 1 and a monoclonal
tumor marker-immunostaining intensity on stored paraffin antibody to cytokeratin 7. Vet Pathol 38:636–643.
slides of breast cancer. J Natl Cancer Inst 88:1054–1059. 50. Ramos-Vara JA, Miller MA, Johnson GC: 2005, Usefulness
31. Jensen TA, Hammond MEH: 2001, The tissue microarray. A of thyroid transcription factor-1 immunohistochemical stain-
technical guide for histologists. J Histotechnol 24:283–287. ing in the differential diagnosis of primary pulmonary tumors
32. Kanai K, Nunoya T, Shibuya K, et al.: 1998, Variation in of dogs. Vet Pathol 42:315–320.
effectiveness of antigen retrieval pretreatment for diagnostic 51. Ramos-Vara JA, Miller MA, Johnson GC, et al.: 2002,
immunohistochemistry. Res Vet Sci 64:57–61. Immunohistochemical detection of thyroid transcription
33. Kiupel M, Webster JD, Kaneene JB, et al.: 2004, The use of factor-1, thyroglobulin, and calcitonin in canine normal,
KIT and tryptase expression patterns as prognostic tools for hyperplastic, and neoplastic thyroid gland. Vet Pathol
canine cutaneous mast cell tumors. Vet Pathol 41:371–377. 39:480–487.
34. Malmstrom PU, Wester K, Vasko J, et al.: 1992, Expression 52. Ramos-Vara JA, Miller MA, Johnson GC, et al.: 2002, Melan
of proliferative cell nuclear antigen (PCNA) in urinary A and S100 protein immunohistochemistry in feline melano-
bladder carcinoma. Evaluation of antigen retrieval methods. mas: 48 cases. Vet Pathol 39:127–132.
Acta Pathol Microbiol Immunol Scand 100:988–992. 53. Ramos-Vara JA, Miller MA, Valli VEO: 2007, Immunohis-
35. Mathews JB, Mason GI: 1984, Influence of decalcifying tochemical detection of multiple myeloma 1/interferon regu-
agents on immunoreactivity of formalin-fixed, paraffin- latory factor 4 (MUM1/IRF-4) in canine plasmacytoma:
embedded tissue. Histochem J 16:771–787. comparison with CD79a and CD20. Vet Pathol 44:875–
36. McNeilly F, Kennedy S, Moffett D, et al.: 1999, A 884.
comparison of in situ hybridization and immunohistochem- 54. Ramos-Vara JA, Saeteele J: 2007, Immunohistochemistry. In:
istry for the detection of a new porcine circovirus in formalin- Making and using antibodies: a practical handbook, ed.
fixed tissues from pigs with post-weaning multisystemic Howard GC, Kaser MR, pp. 273–314. CRC Press, Boca
wasting syndrome (PMWS). J Virol Meth 80:123–128. Raton, FL.
37. Morgan JM, Navabi H, Jasani B: 1997, Role of calcium 55. Rasmussen OF: 2006, Controls. In: Immunohistochemical
chelation in high-temperature antigen retrieval at different pH staining methods, ed. Key M, 4th ed., pp. 113–118. DAKO
values. J Pathol 182:233–237. Corporation, Carpinteria, CA.
38. Olapade-Olaopa E, Oluwabunmi DM, Ogunbiyi J, et al.: 56. Reid V, Doherty J, McIntosh G, et al.: 2007, The first
2001, Further characterization of storage-related alterations in quantitative comparison of immunohistochemical rabbit and
immunoreactivity of archival tissue sections and its implica-
mouse monoclonal antibody affinities using Biacore analysis.
tions for collaborative multicenter immunohistochemical
J Histotechnol 30:177–182.
studies. Appl Immunohistochem Mol Morphol 9:261–266.
57. Rhodes A, Jasani B, Couturier J, et al.: 2002, A formalin-
39. Packeisen J, Buerger H, Krech R, Boecker W: 2002, Tissue
fixed, paraffin-processed cell line standard for quality control
microarrays: a new approach for quality control in immuno-
of immunohistochemical assay of HER-2/neu expression in
histochemistry. J Clin Pathol 55:613–615.
breast cancer. Am J Clin Pathol 117:81–89.
40. Packeisen J, Korsching E, Herbst H, et al.: 2003, Demysti-
58. Rüdiger T, Höfler H, Kreipe HH, et al.: 2002, Quality
fied…tissue microarray technology. J Clin Pathol 56:198–204.
assurance in immunohistochemistry. Results of interlabora-
41. Pileri SA, Roncador G, Ceccarelli C, et al.: 1997, Antigen
tory trial involving 172 pathologists. Am J Surg Pathol
retrieval techniques in immunohistochemistry: comparison of
26:873–882.
different methods. J Pathol 183:116–123.
42. Prioleau J, Schnitt SI: 1995, p53 antigen loss in stored paraffin 59. Sabattini E, Bisgaard K, Ascani S, et al.: 1998, The
slides. N Engl J Med 332:1521–1522. EnVision+TMsystem: a new immunohistochemical method
43. Ramos-Vara JA: 2005, Technical aspects of immunohisto- for diagnostics and research. Critical comparison with the
chemistry. Vet Pathol 42:405–426. APAAP, ChemMateTM, CSA, LABC, and SABC techniques.
44. Ramos-Vara JA, Beissenherz ME: 2000, Optimization of J Clin Pathol 51:506–511.
immunohistochemical methods using two different antigen 60. Schweitzer B, Wiltshire S, Lambert J, et al.: 2000, Immuno-
retrieval methods on formalin-fixed, paraffin-embedded tis- assays with rolling circle DNA amplification: antigen detec-
sues: experience with 63 markers. J Vet Diagn Invest tion. Proc Natl Acad Sci USA 97:10113–10119.
12:307–311. 61. Seidal T, Balaton AJ, Battifora H: 2001, Interpretation and
45. Ramos-Vara JA, Beissenherz ME, Miller MA, et al.: 2000, quantification of immunostains. Am J Surg Pathol 25:1204–
Retrospective study of 338 canine oral melanomas with 1207.
clinical, histologic, and immunohistochemical review of 129 62. Shi SR, Cote RJ, Taylor CR: 2001, Antigen retrieval
cases. Vet Pathol 37:597–608. techniques: current perspectives. J Histochem Cytochem
46. Ramos-Vara JA, Beissenherz ME, Miller MA, et al.: 2001, 49:931–937.
Immunoreactivity of A103, an antibody to Melan A, in canine 63. Shi SR, Key ME, Kalra KL: 1991, Antigen retrieval in
steroid-producing tissues and their tumors. J Vet Diagn Invest formalin-fixed, paraffin-embedded tissues: an enhancement
13:328–332. method for immunohistochemical staining based on micro-
47. Ramos-Vara JA, Kiupel M, Miller MA: 2005, Veterinary wave oven heating of tissue sections. J Histochem Cytochem
diagnostic immunohistochemistry: a survey of 47 laboratories. 39:741–748.
J Histotechnol 28:19–23. 64. Taylor CR, Shi S-R, Barr NJ, et al.: 2006, Techniques of
48. Ramos-Vara JA, Miller MA, Boucher M, et al.: 2003, immunohistochemistry: principles, pitfalls, and standardiza-
Immunohistochemical detection of uroplakin III, cytokeratin tion. In: Diagnostic immunohistochemistry, ed. Dabbs DJ,
7, and cytokeratin 20 in canine urothelial tumors. Vet Pathol 2nd ed., pp. 1–42. Churchill Livingstone, Philadelphia, PA.
40:55–62. 65. Thrustfield M: 1995. Veterinary epidemiology, 2nd ed.
49. Ramos-Vara JA, Miller MA, Johnson GC: 2001, Immuno- Blackwell Science, Oxford, United Kingdom.
histochemical characterization of canine hyperplastic hepatic 66. True LD: 1990. Atlas of diagnostic immunohistopathology.
lesions and hepatocellular and biliary neoplasms with Lippincott Williams & Wilkins, Philadelphia, PA.
Immunohistochemical techniques in veterinary diagnostic laboratories 413

67. Tubbs RR, Nagle R, Leslie K, et al.: 1998, Extension of useful p53 protein and MIB1 antigen. Appl Immunohistochem Mol
reagent shelf life beyond manufacturer’s recommendations. Morphol 8:61–70.
Arch Pathol Lab Med 122:1051–1052. 73. Wetherington RW, Cooper HS, Al-Saleem T, et al.: 2002,
68. Van Hecke D: 2002, Routine immunohistochemical staining Clinical significance of performing immunohistochemistry on
today: choices to make, challenges to take. J Histotechnol cases with a previous diagnosis of cancer coming to a national
25:45–54. comprehensive cancer center for treatment or second opinion.
69. Vilches-Moure JG, Ramos-Vara JA: 2005, Comparison of Am J Surg Pathol 26:1222–1230.
rabbit monoclonal and mouse monoclonal antibodies in 74. Wick MR, Mills SE: 2001, Consensual interpretive guidelines
immunohistochemistry in canine tissues. J Vet Diagn Invest for diagnostic immunohistochemistry. Am J Surg Pathol
17:346–350. 25:1208–1210.
70. Vis AN, Kranse R, Nigg AL, et al.: 2000, Quantitative 75. Wiedorn KH, Goldmann T, Henne C, et al.: 2001, EnVision+,
analysis of the decay of immunoreactivity in stored prostate a new dextran polymer-based signal enhancement technique
needle biopsy sections. Am J Clin Pathol 113:369–373. for in situ hybridization (ISH). J Histochem Cytochem
71. Werner M, Chott A, Fabiano A, Battifora H: 2000, Effect of 49:1067–1071.
formalin tissue fixation and processing on immunohistochem- 76. Yaziji H, Barry T: 2006, Diagnostic immunohistochemistry:
istry. Am J Surg Pathol 24:1016–1019. what can go wrong? Adv Anat Pathol 13:238–246.
72. Wester K, Wahlund E, Sundstrom C, et al.: 2000, Paraffin 77. Zeheb R: 2006, Automating immunohistochemistry. In:
section storage and immunohistochemistry: effects of time, Immunohistochemical staining methods, ed. Key M, 4th ed.,
temperature, fixation, and retrieval protocol with emphasis on pp. 103–106. DAKO Corporation, Carpinteria, CA.

You might also like